You are on page 1of 18

Immunity

Review

Cancer-Cell-Intrinsic Mechanisms
Shaping the Tumor Immune Landscape
Max D. Wellenstein1 and Karin E. de Visser1,*
1Division of Tumor Biology & Immunology, Oncode Institute, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands

*Correspondence: k.d.visser@nki.nl
https://doi.org/10.1016/j.immuni.2018.03.004

Owing to their tremendous diversity and plasticity, immune cells exert multifaceted functions in tumor-
bearing hosts, ranging from anti-tumor to pro-tumor activities. Tumor immune landscapes differ greatly be-
tween and within cancer types. Emerging evidence suggests that genetic aberrations in cancer cells dictate
the immune contexture of tumors. Here, we review the current understanding of the mechanisms whereby
common drivers of tumorigenesis modulate the tumor immune milieu. We discuss these findings in the
context of clinical observations and examine how cancer-cell-intrinsic properties can be exploited to maxi-
mize the benefit of immunomodulatory therapies. Understanding the relationship between cancer cell-
intrinsic genetic events and the immune response may enable personalized immune intervention strategies
for cancer patients.

Introduction cancer cell-extrinsic regulators of cancer biology, the immune


The recognition of cancer as a genetic disease is more than a system.
century old and stems from observations by David von Hanse- Similar to its physiological function, the immune system exerts
mann and Theodor Boveri that cancer cells display chromo- multifaceted tasks in tumor-bearing hosts, with different immune
somal abnormalities (Boveri, 1914; von Hansemann, 1890). In cells playing different and sometimes opposing roles. The
the early 20th century, Francis Rous revealed that retroviruses composition and function of immune cells in tumors differs
could drive sarcoma formation in chickens (Rous, 1911). Many greatly between, but also within, cancer types. For example, of
decades later, in 1970, the Rous sarcoma virus was found to the breast cancer subtypes, triple-negative breast cancer
carry a gene called v-Src, the first oncogene to be identified (TNBC) presents with highest levels of tumor-infiltrating lympho-
(Duesberg and Vogt, 1970; Stehelin et al., 1976). Concurrently, cytes (TIL) and macrophages (Medrek et al., 2012; Stanton et al.,
it was discovered that not only activation, but also inactivation 2016). Striking differences in relative leukocyte composition be-
of so-called tumor suppressor genes (TSGs) can lead to tumor- tween different tumor types were observed in a study that inte-
igenesis (Knudson, 1971). (Proto-)oncogenes and TSGs regulate grated gene expression and clinical outcome data of over
essential cellular processes like cell cycle, apoptosis, migration, 18,000 human tumors (Gentles et al., 2015). Moreover, this study
and survival, and genetic aberrations that lead to dysregulation revealed considerable variation in intratumoral presence of
or loss of function of these genes can result in malignant trans- certain immune cell subsets and how these were associated
formation. The generation of transgenic mice carrying an acti- with cancer-specific outcomes. For example, whereas memory
vated oncogene, also called oncomice, in the 1980s and TSG CD4+ T cells were associated with adverse outcome in bladder
knockout mice in the 1990s further substantiated the notion cancer patients, they correlated with favorable outcome in lung
that oncogene expression or loss of TSGs in normal mammalian adenocarcinoma patients (Gentles et al., 2015), suggesting
cells leads to cancer development (Adams et al., 1985; Done- that differences in immune profile are not only phenotypically
hower et al., 1992; Hanahan et al., 2007; Stewart et al., 1984). distinct but are also of functional consequence. But what deter-
The dependency of cancers on these dysregulated genes mines this substantial variation in immune contexture between
was demonstrated in genetically engineered mouse models different tumors? Given the surge of interest in utilizing immuno-
(GEMMs) in which de-activation of oncogenes or re-expression modulatory drugs for the treatment of cancer patients, it is crit-
of TSGs in fully established tumors led to rapid tumor regression ical to understand the underlying tumor characteristics that
(Fisher et al., 2001; Jain et al., 2002; Moody et al., 2002; Ventura dictate the inter-tumor heterogeneity in immune landscapes
et al., 2007). These insights into the causal role of genetic aber- and to use this knowledge for rational decision-making in the
rations in cancer initiation and progression spurred the long-held clinical use of immunomodulatory strategies.
belief that tumorigenesis is entirely driven by cancer-cell-intrinsic In this review, we will discuss recent insights into how cancer
genetic traits. However, over the past couple of decades, this cell-intrinsic properties can dictate the immune landscape of tu-
dogma has been challenged by new experimental evidence mor-bearing hosts. Specifically, we will examine which genetic
demonstrating that genetic aberrations alone are required, but aberrations correlate with immune cell composition in human
not sufficient, for a cancer to develop. Like a seed needing fertile tumors. Next, we will discuss the current knowledge on onco-
soil for successful germination, cancer cells only survive and gene- and TSG-dependent signaling pathways that underlie
develop into invasive tumors in an environment that provides the differential crosstalk of cancer cells with the immune system
sufficient nutrients and oxygen, and that lacks strong cytotoxic as identified in genetically engineered mouse tumor models
signals. In this review, we will focus on one of the most influential (GEMMs). Finally, we will discuss how the growing insights into

Immunity 48, March 20, 2018 ª 2018 Elsevier Inc. 399


Immunity

Review

these mechanisms may open new avenues for personalized im- there is a substantial number of patients with good response
mune intervention strategies for cancer patients. and low mutational load and vice versa (Balli et al., 2017;
Charoentong et al., 2017; Hugo et al., 2016; Rizvi et al., 2015;
Genetic Makeup Influencing the Immune Contexture of Robinson et al., 2017; Spranger et al., 2016). These observations
Tumors—Observations from the Clinic suggest that for some tumors the mutational burden of tumors
In 1863, the German pathologist Rudolf Virchow was the first to can serve as a quantitative measure for T cell abundance and
hypothesize a link between the development of tumors and the likelihood to respond to immune checkpoint inhibitors. However,
inflammatory state of their anatomical location (Balkwill and there are clearly additional determinants of the immune contex-
Mantovani, 2001). Around the same time, William Coley, pioneer ture in tumors besides mutational load.
of cancer immunotherapy, demonstrated that some patients Distinct molecular subtypes of human cancers can be associ-
displayed tumor regression after being injected with immune ated with a defined immune composition and activation state in
stimulatory Streptococcus pyogenes cultures (Coley, 1893). the tumor microenvironment. Several cancer types can be
Nowadays, it is fully established that inflammation can be caus- subtyped based on their molecular and genetic profile, thus
ally linked with human cancers and that the immune infiltrate of forming separate classes within a given tumor type, often with
human tumors contains prognostic and predictive information distinct progression characteristics and treatment regimens.
(Diakos et al., 2014; Gentles et al., 2015). Moreover, cancer For example, breast tumors can be classified as Luminal A
immunotherapy has revolutionized cancer treatment (Yang, (ER/PR+, HER2–), Luminal B (ER/PR+, HER2+/–), HER2-enriched
2015), illustrating that immune cells can be harnessed success- (HER2+), and triple-negative/basal-like (ER/PR/HER2–) (Parker
fully to destroy tumors in a proportion of cancer patients. et al., 2009). It has been reported that CD8+ T cells preferentially
Recently, studies have started to explore the cancer cell charac- infiltrate in triple negative tumors and those patients with high in-
teristics—including the genetic makeup—that play a critical role tratumoral T cell abundance show better disease-free survival
in dictating the heterogeneity in immune landscape between (Chen et al., 2014; Medrek et al., 2012; Savas et al., 2016; Stan-
different tumors. Studies aimed at assessing the link between ton et al., 2016). Breast tumors that express hormone receptors
the genetics of human tumors and the immune infiltrate can be or HER2 are more frequently infiltrated by FoxP3+ regulatory
roughly divided into three categories: (1) studies that have as- T cells (Tregs) compared to other subtypes, suggesting depen-
sessed the extent of the mutational load of tumors with T cell dency on these receptors in the establishment of an immuno-
abundance, specificity and activity, (2) studies that have linked suppressive milieu (Decker et al., 2012; Jiang et al., 2015).
distinct molecular tumor subtypes with a certain immune land- Accordingly, the presence of Tregs in breast tumors predicted
scape, and (3) studies that have focused on the association be- metastatic progression and poor survival (Jiang et al., 2015;
tween defined oncogenic driver mutations or loss of TSGs and Liu et al., 2011). For other cancer types, such as colorectal can-
parameters of the inflammatory tumor microenvironment. In cer, glioblastoma, and head and neck cancer, similar subtype-
this section, we will discuss the findings of these three different specific tumor immune infiltrates have been observed (Becht
strategies to assess the impact of genetic events on the cross- et al., 2016; Doucette et al., 2013; Keck et al., 2015; Wang
talk with the immune system. et al., 2017) (Table 1). These clinical observations indicate that
The core function of the adaptive immune system is to recog- different molecular subtypes of tumors can be characterized
nize and destroy cells expressing non-self-antigens, while not by distinct immune landscapes. However, due to the complex
responding to self-antigens. Because cancers arise from host nature that underlies molecular subtypes, the exact genes and
cells, these cancer cells, with the exception of viral-associated mechanisms that determine this immune heterogeneity cannot
cancers, do not express the typical immunogenic foreign anti- be distilled from these studies.
gens as seen in infections. The recent clinical breakthrough of A growing body of clinical observations indicates that defined
immune checkpoint inhibitors has fueled studies aimed at iden- oncogenic driver mutations and loss of TSGs in human cancers
tifying the tumor antigens that are recognized by effective anti- are also correlated with changes in immune composition and
tumor immune responses. This resulted in the hypothesis that immunotherapy response. For example, loss of NF1 in glioblas-
a higher mutational load of a tumor will inevitably result in more tomas associated with an increase in macrophages in the tumor
‘‘foreign’’ peptide presentation and consequently higher immu- (Wang et al., 2017). Another study showed that loss of heterozy-
nogenicity of the tumor. Mutations and other genomic rearrange- gosity (LOH) or mutation of TP53 in ER-negative and basal-like
ments in cancer cells can encode for neo-antigens, antigens breast tumors is associated with decreased intratumoral expres-
uniquely expressed by the tumor, that when presented by sion of a cytotoxic T cell signature and poor survival (Quigley
MHC molecules can potentially be recognized by the endoge- et al., 2015). These studies indicate that a single TSG can be
nous T cell repertoire (Schumacher and Schreiber, 2015). associated with the immune composition of the tumor, across
Indeed, neo-antigen-specific T cells have been observed in mel- different tumor subtypes, and therefore might be a dominant
anoma patients (Lennerz et al., 2005; Linnemann et al., 2015; driving force of immune influx. Furthermore, in pancreatic ductal
Robbins et al., 2013; van Rooij et al., 2013; Wölfel et al., 1995) adenocarcinoma (PDAC), expression of genes associated with
and tumor types with a relatively high mutational burden, such cytotoxic T cell function and immune checkpoint molecules
as melanoma, non-small cell lung cancer (NSCLC) and microsat- was inversely linked with amplification of MYC, NOTCH2, and
ellite-instable (MSI) tumors display increased T cell influx and FGFR1, but not with mutational load (Balli et al., 2017). The
have an overall better response rate to immunotherapeutics reduced expression of cytolytic immune response markers in
compared to tumors with a lower mutational load (Le et al., these MYC-, NOTCH2-, and FGFR1-amplified tumors was
2015; Rizvi et al., 2015; Van Allen et al., 2015). Nevertheless, observed across the different PDAC subtypes (Bailey et al.,

400 Immunity 48, March 20, 2018


Immunity

Review

Table 1. Clinical Observations on Tumor Subtype and Genotype-Immunophenotype Relations


Determinant of tumor immune Effect on therapy/disease
landscape Cancer type Immune cell subset outcome Reference
Tumor subtype
CMS1 CRC [ Cytotoxic T cellsa Overall favorable response (Becht et al., 2016)
to immune checkpoint blockade
Mesenchymal Glioblastoma [ Immunosuppressive NA (Doucette et al., 2013)
cellsa
[ T effector cellsa
[ Macrophages, (Wang et al., 2017)
neutrophilsa
Triple-negative/basal-like Breast cancer [ CD8+ T cells, High CD8+ T cell abundance (Chen et al., 2014; Medrek
macrophages gives high overall survival et al., 2012; Savas et al.,
2016; Stanton et al., 2016)
ER/PR/HER2+ [ Tregs High Treg abundance gives (Decker et al., 2012; Jiang
poor overall survival et al., 2015; Liu et al., 2011)
Inflamed/mesenchymal HPV+/– HNSCC [ CD8+ T cellsa NA (Keck et al., 2015)
Mutated oncogenes or tumor suppressor gene
TP53 loss or mutation ER– & basal-like Y Cytotoxic T cellsa Poor survival (Quigley et al., 2015)
breast cancer
Pan-cancer Y Cytotoxic T, NK cellsa NA (Rooney et al., 2015)
MYC, NOTCH2, FGFR1 PDAC Y Cytotoxic T cellsa NA (Balli et al., 2017)
amplification
MYC amplification Neuroblastoma Y T cellsa NA (Layer et al., 2017)

PIK3CA, MET mutations Pan-cancer [ Cytotoxic T, NK cellsa NA (Rooney et al., 2015)


BRAF mutations Thyroid cancer [ Immunosuppressive NA (Charoentong et al., 2017)
cellsa
RAS mutations [ T cellsa
VHL, STK11 mutations Pan-cancer Y Macrophagesa NA (Rooney et al., 2015)
NF1 loss Glioblastoma [ Macrophages NA (Wang et al., 2017)
Abbreviations: CRC, colorectal cancer. HNSCC, head and neck squamous cell carcinoma. PDAC, pancreatic ductal adenocarcinoma. NA, Not
assessed.
a
Immune cell composition based on gene expression signatures.

2016; Balli et al., 2017) and suggests that aberrant expression of JAK1/2 (Shin et al., 2017). Using human melanoma cell lines, it
oncogenic pathways also dominantly impacts the composition was shown that JAK1/2 mutations led to an impaired IFN
of the pancreatic tumor microenvironment (Table 1). signaling pathway-mediated PD-L1 expression, suggesting
Genetic aberrations in tumors can also influence the T cell that also JAK-STAT signaling is involved in regulating immune
response by altering expression levels of immune checkpoint checkpoint expression. These findings indicate that screening
molecules by cancer cells. In a cohort of lung adenocarcinoma for expression of certain oncogenes or loss of function of specific
patients, accumulation of p53 in tumor cells, which is indicative TSGs might be exploited to improve the stratification of cancer
of mutations in TP53, correlated with increased PD-L1 expres- patients for therapeutic targeting the PD-1/PD-L1 axis.
sion, while mutant EGFR tumors were characterized by low The link between the genetic makeup of tumors and their im-
expression of PD-L1 (Cha et al., 2016). In contrast, another study mune contexture was further strengthened by recent high-
showed that EGFR mutated lung tumors have high levels of PD- throughput next generation sequencing (NGS) studies, which
L1 (Akbay et al., 2013), demonstrating that the role of mutant allow an unbiased assessment of the genetics of tumors in par-
EGFR in regulating PD-L1 expression is still under debate. In allel with high-resolution mapping of the tumor immune land-
metastatic neuroblastoma, amplification of MYCN correlated scape. By correlating an RNA-based metric of immune cytolytic
with low expression of PD-L1 and a reduced T cell gene-expres- activity (mainly associated with T and NK cell function) with ge-
sion signature in the tumor compared to MYCN-normal tumors netic data from the Cancer Genome Atlas (TCGA) dataset, it
(Layer et al., 2017). Moreover, MYCN overexpression inversely was shown that immune activity varies substantially across tu-
correlated with natural killer (NK) cell-activating factors such as mor types (Rooney et al., 2015). Consistent with the concept
NKG2D in primary human neuroblastoma cell lines (Brandetti that a higher mutational load increases tumor immunogenicity,
et al., 2017). In addition, resistance to anti-PD-1 treatment in there was a positive correlation between adaptive immune acti-
melanoma and MSI CRC patients correlated with mutations in vation gene signatures and mutational load across tumor types

Immunity 48, March 20, 2018 401


Immunity

Review

(Rooney et al., 2015). Interestingly, this study also revealed that (Figure 1, Table 2). One notable example is NF-kB, a transcrip-
expression of genes associated with cytotoxic immune activa- tion factor that controls cell survival and proliferation, but also
tion was elevated in tumors with mutations in PIK3CA or MET, production of inflammatory cytokines. For example, NF-kB
while TP53 mutant tumors displayed low levels of these genes signaling promoted tumor development in the KrasLSL-G12D/+;
(Rooney et al., 2015). Additionally, mutations in VHL and Trp53F/F lung adenocarcinoma model (Meylan et al., 2009). Inter-
STK11 associated with reduced macrophage signatures (Roo- estingly, NF-kB activity was increased upon loss of p53, and
ney et al., 2015). In another study into genotype-immunopheno- restoration of p53 expression reduced its activity. Cancer cell-
type relationships, it was found that BRAF-mutated thyroid intrinsic NF-kB inactivation resulted in increased intratumoral
tumors were characterized by infiltration of immunosuppressive immune cell influx and impaired lung cancer formation in
cells, while the RAS-mutated subtype contained higher T cell KrasLSL-G12D;Trp53F/F mice (Meylan et al., 2009), showing a link
influx and displayed downregulation of MHC molecules, between loss of p53, NF-kB pathway activation, and an inflam-
despite comparable mutational load (Charoentong et al., 2017). matory tumor microenvironment. As one of the most frequently
Accordingly, oncogenic mutations also link with response to mutated genes in cancer (Kastenhuber and Lowe, 2017), the tu-
immunotherapy. Using human datasets to predict response to mor suppressor p53 can potentially regulate the immune infil-
anti-CTLA-4 therapy in melanoma patients, it was demonstrated trate in a wide variety of tumor types, through its interactions
that mutations in oncogenes such as KRAS, ATM, and mTOR with NF-kB or otherwise. Indeed, the control of the pro-inflam-
correlated with good immunotherapy response for some tumor matory NF-kB pathway by p53 appears to be occurring across
types (Ock et al., 2017). These studies demonstrate that NGS cancer types (Cooks et al., 2014). For example, in the Pgr-
studies can reveal relationships between cancer-associated cre;Cdh1F/F;Trp53F/F mouse model for endometrial cancer, the
genes, activation of immune cells and response to immunother- combined loss of E-cadherin and p53 resulted in increased
apies in a high-throughput and high-resolution manner. NF-kB activity, which correlated with elevated cytokine expres-
Together, these observations suggest that mutational load, tu- sion and increased influx of macrophages, as compared to dele-
mor subtype, and aberrant expression of oncogenes and TSGs tion of either gene alone (Stodden et al., 2015). However, in
highly impact the tumor microenvironment. Interestingly, for another mouse model in which endometrial tumorigenesis is
certain tumors, the tumor driver genes, mutational load, and sub- driven by loss of PTEN, loss of p53 did not alter neutrophil influx
type are intrinsically linked, as for example aberrant expression into early lesions (Blaisdell et al., 2015), suggesting that this ef-
of BRCA1 impairs the DNA damage repair machinery and there- fect might be context dependent. Together, these and other
fore has consequences for the mutational load of a tumor. studies show that NF-kB, key regulator of immune signaling in
However powerful, these genotype-immunophenotype studies the tumor microenvironment, is controlled by p53. In several tu-
in human cancers leave several questions open. Due to the mor models, loss of p53 activates the NF-kB pathway, stimu-
descriptive nature of these analyses, these studies do not yield lates the production of cytokines and other pro-inflammatory
mechanistic insights into causal relationships between tumor mediators from cancer cells, which through paracrine interac-
genetics and the immune composition. From a therapeutic tions modify the immune contexture.
perspective, it is important to assess whether a causal link be- Studies in mouse models in which chemical-induced inflam-
tween tumor genetics and immune contexture exists and to mation drives malignant conversion and progression show that
elucidate the underlying molecular mechanisms, since this the NF-kB-mediated inflammatory response can also be a
would open new avenues for personalized immune intervention driving force of tumorigenesis in p53-knockout models. For
strategies. Of note, the above described clinical studies often example, azoxymethane (AOM)-induced colonic tumorigenesis
rely on the analysis of a small tumor biopsy at a given time point, was enhanced in Villin-cre;Trp53F/F mice that harbor p53 dele-
and therefore may overlook intratumoral heterogeneity and tu- tion in intestinal epithelial cells, as compared to mice with p53
mor evolution. For these reasons, mechanistic studies in relevant proficient intestinal epithelial cells (Schwitalla et al., 2013). Mech-
GEMMs that mimic the development, heterogeneity and pro- anistic studies in these mice revealed that loss of p53 impaired
gression of human tumors in an immune-proficient setting are the removal of pre-neoplastic transformed cells and induced
key to understand how cancer cell-intrinsic properties can NFkB-dependent cytokine production, thus driving an inflamma-
dictate the tumor immune landscape (Kersten et al., 2017). In tory tumor microenvironment (Schwitalla et al., 2013). Impor-
the next sections, we will discuss recent insights into these tantly, genetic ablation of IKKb, a protein involved in NF-kB
mechanisms and how these insights can be translated into activation, in cancer cells or myeloid cells, reduced tumor prolif-
personalized immune intervention strategies. Given the growing eration and invasion, demonstrating that NF-kB signaling in p53
interest in the role of the immune system in tumorigenesis, we null cancer cells or in surrounding myeloid cells plays a funda-
anticipate that more pathways will be uncovered in the years mental role in tumor progression (Schwitalla et al., 2013).
to come. A critical feature of p53 biology in cancer not addressed in
these studies is its wide variety of both activating and inactivat-
NF-kB and p53: Central Nodes in Cancer Cell-Mediated ing mutations, leading to very diverse and sometimes even
Changes in the Inflammatory Microenvironment opposing functions (Muller and Vousden, 2014). How one of
The mechanisms by which oncogenes and TSGs orchestrate these p53 mutations affects NF-kB activation, was addressed
the inflammatory tumor microenvironment are now being uncov- in a gain-of-function (GOF) mutant p53G515A mouse model that
ered. Specific cancer-associated genes, besides driving cancer was repeatedly exposed to dextran sodium sulfate (DSS) to stim-
cell-intrinsic programs, also change the secretome of cancer ulate colitis-induced colorectal cancer (CRC) (Cooks et al.,
cells and thereby change the immune microenvironment 2013). Repair of DSS-induced damaged tissue was impaired in

402 Immunity 48, March 20, 2018


Immunity

Review
Figure 1. Cancer Cell-Intrinsic Signaling
A B C Pathways that Shape the Tumor Immune
Landscape
(A) The p53 pathway can modulate the immune
microenvironment of the tumor by regulating NF-kB
signaling, that is generally activated by loss or loss-
of-function (LOF) mutation of p53. This results in
increased cytokine production by tumor cells and
recruitment and activation of immune cells, such as
macrophages (Cooks et al., 2013; Schwitalla et al.,
2013). In addition, by activating ROS, mutant p53
can induce JAK-STAT signaling and thereby in-
crease macrophage, neutrophil, and CD4+ T cell
frequencies in the tumor, while concurrently
reducing CD8+ T cell levels (Wörmann et al., 2016).
D (B) Mutant KRAS can increase GM-CSF by can-
cer cells and thereby promote neutrophil recruit-
F ment to the tumor (Pylayeva-Gupta et al., 2012).
(C) Activated Notch signaling can signal to
E monocytes and macrophages by driving CCL2
G and IL-1b expression. Notch also drives TGF-b
receptor and uPA expression, of which the latter
is involved in activating macrophage-derived
TGF-b, thus inducing a growth promoting
H K signaling loop (Shen et al., 2017). Notch can also
I J limit the anti-tumor immune response by inhibiting
C/EBPb and thereby limiting expression of IL-1,
IL-6, and IL-8 (Hoare et al., 2016).
(D) Loss of tumor suppressor gene LKB1 can
drive production of G-CSF, CXCL7, and IL-6 by
the tumor, which promotes neutrophil recruit-
ment, which can block anti-tumoral cytotoxic
T cells (Koyama et al., 2016).
(E) ZBTB7a blocks CXCL5 production by binding
its promoter, and loss of ZBTB7a therefore can
lead to CXCL5-mediated neutrophils recruitment
(Bezzi et al., 2018).
(F) High mutational load in tumors can increase
the number of neo-antigens and thus potentially
increase neo-antigen-specific T cell responses.
(G) PTEN can negatively regulate NF-kB signaling. Therefore, loss of PTEN increases NF-kB-mediated expression of cytokines and growth factors that drive
macrophage, neutrophil, and Treg accumulation in the tumor (Ying et al., 2011).
(H) MYC can regulate macrophage recruitment, which is promoted by p53 loss (Yetil et al., 2015). Additionally, by inducing CCL5 and IL-1b, MYC can promote
mast cell recruitment and activation (Shchors et al., 2006; Soucek et al., 2007). MYC can also induce CCL9 and IL-23 expression, the former of which induces
macrophage recruitment, while the latter limits NK, T, and B cell accumulation in the tumor (Kortlever et al., 2017). MYC can also inhibit CD4+ T cells and
macrophages by regulating PD-L1 and CD47 expression on tumor cells (Casey et al., 2016). Lastly, the anti-tumor NK- and CD8+ T cell-response to MYC
amplified tumors can be counteracted by additional loss of p53 in the tumor, while amplification of Bcl-2 promotes anti-tumor immunity (Schuster et al., 2011).
(I) SMAD4 can suppress YAP1 signaling, and loss of SMAD4 in tumors therefore drives YAP1-mediated CXCL5 production, which recruits immunosuppressive
neutrophils (Wang et al., 2016).
(J) PRKCI amplification can also induce YAP signaling. Activation of YAP1 here induces TNFa-mediated recruitment and activation of immunosuppressive
neutrophils (Sarkar et al., 2017).
(K) Activated Wnt signaling via b-catenin can limit the priming of CD8+ T cells by suppression of CCL4 production, which would otherwise activate CD103+ DCs
(Spranger et al., 2015).

p53G515A mice. Combined with enhanced NF-kB activity and fiencient tumors displayed similar STAT3-dependent immune
extended inflammation, this led to an increase in colorectal tu- evasion and accelerated tumor growth, which both could be
mor incidence in mice (Cooks et al., 2013). In addition, reversed by pharmacological targeting of JAK-STAT signaling
p53G515A mutant intestinal organoids derived from these mice (Wörmann et al., 2016). These findings indicate that different mu-
showed increased tumor necrosis factor alpha (TNF-a) and tations of p53 can shape the tumor microenvironment in a
CXCL1 production when compared to p53–/– cells, which could distinct manner. In future studies, it would be interesting to sys-
be reverted by NF-kB knockdown (Cooks et al., 2013). In line tematically dissect the differences between gain- and loss-of-
with these experimental findings, expression of mutant TP53 function p53 mutations on NF-kB interactions and the immune
correlated with NF-kB expression in human CRC patients landscape of the tumor. Altogether, these studies demonstrate
(Cooks et al., 2013). These findings show that this GOF mutant the profound role of p53-mediated regulation of key immune
p53 induces aberrant NF-kB interactions, leading to different in- signaling pathways such as NF-kB and STAT signaling, and its
flammatory phenotypes than observed after loss of p53. In a downstream effects on the tumor immune landscape.
mouse model for pancreatic cancer, p53R172H has been re-
ported to elicit similar immune phenotypes as loss of p53. MYC: A Key Controller of the Immune Microenvironment
KrasG12D;p53R172H mutant mouse pancreatic tumors drive in- The MYC oncogene is one of the most frequently amplified onco-
flammatory responses via ROS and JAK2-STAT3 activation genes in several tumor types, such as lymphoma, breast cancer,
(Wörmann et al., 2016). Here, both p53R172H mutant and p53-de- and NSCLC (Beroukhim et al., 2010). As a transcription factor,

Immunity 48, March 20, 2018 403


404 Immunity 48, March 20, 2018

Table 2. Genetic Aberrations Influencing the Immune Landscape of Tumors


Genetic Consequence for
Gene aberration intratumoral immune cells Signaling involved Tumor type Tumor model Reference
AKT Loss Macrophages Y AKT deletion decreases tumorigenesis by reducing Liver cancer Alb-cre;PtenF/F and (Debebe et al., 2017)
pro-tumorigenic Wnt-producing macrophages in Alb-cre;PtenF/F;Akt2F/F
the tumor
ATR Deletion Macrophages [ NA Melanoma Tyr::ERT2;BrafV600E/+;PtenF/F (Chen et al., 2017)
B cells [, CD8 T cells Y
+ and Tyr::ERT2;BrafV600E/+;
PtenF/F;ATRF/F
b catenin Amplification CD8+ T cells Y Active b-catenin inhibits CCL4, thus inhibiting Melanoma Tyr:cre-ER;BrafLSL-V600E/+; (Spranger et al., 2015)
CD8+ T cell priming by CD103+ DCs. PtenF/F and Tyr:cre-ER;
BrafLSL-V600E/+;PtenF/F;
LSL-CAT-STA
CKIa Loss Macrophages Y Loss of CKIa triggers an inflammatory SASP. CRC Villin-cre;CKIaF/F, Villin-cre; (Pribluda et al., 2013)
Subsequent loss of p53 or p21 leads to CKIaF/F;p21 / and Villin-
inflammation-accelerated tumorigenesis. cre;CKIaF/F;Trp53F/F
EGFR Mutation Macrophages, neutrophils [ NA NSCLC Ccsp-rtTA;TetO-EgfrL858R (Busch et al., 2016)
CD8 T cells Y
+

CD8+ T cells Y EGFR pathway activates PDL1 expression in Ccsp-rtTA;TetO-EGFRT790M, (Akbay et al., 2013)
bronchial epithelial cells EGFRT790M/L858R and
EGFRexon 19 del/T790M
FAK Amplification Macrophages, neutrophils, Potentially via STAT3 signaling. PDAC p48-Cre;KrasLSL-G12D/+ (Jiang et al., 2016)
monocytes [
CD3+ T cells Y, Tregs [ Potentially due to immunosuppressive
myeloid cells
FGFR Activation Neutrophils [ FGFR drives mTOR signaling, which causes Breast cancer MMTV-Wnt1, MMTV-Wnt1- (Welte et al., 2016)
increase in G-CSF production, driving neutrophil iFGFR and MMTV-cre;
expansion, thus promoting tumor progression Trp53F/F;PtenF/F
IFNAR1 Mutation NK cells Y, neutrophils [ Inactivating mutant of IFNAR1 promotes the CRC AOM-DSS induced (Katlinski et al., 2017)
CD8+ cells Y establishment of an immunosuppressive
microenvironment and tumor progression
KRAS Mutation Myeloid cells [ NA NSCLC KrasLSL-G12D/+ and (Busch et al., 2016)
+
T cells (CD8 , Treg, gd KrasLSL-G12D/+;Trp53F/F
T cells) [
LKB1 Loss Neutrophils [, Macrophages, Loss of Lkb1 leads to an increase in CXCL7, NSCLC KrasLSL-G12D/+ and (Koyama et al., 2016)
CD4+, CD8+ T cells Y G-CSF and IL-6, which drive neutrophil increase. KrasLSL-G12D/+;Lkb1F/F
Neutrophils decrease IFNg+ T cells in the tumor.

Review
mTOR Amplification NK cells, macrophages [ mTOR regulates IL-1a levels, and IL-1a activates Liver cancer Hydrodynamic tail-vein (Herranz et al., 2015;
NFkB, thus driving SASP and immune cell injection of NRASG12V Laberge et al., 2015)
recruitment
T, B cells [ mTOR activates tumor suppressive SASP

Immunity
(Continued on next page)
Review
Immunity
Table 2. Continued
Genetic Consequence for
Gene aberration intratumoral immune cells Signaling involved Tumor type Tumor model Reference
MYC Loss Macrophages, neutrophils Y NA Pancreatic cancer RIP1-Tag2 and TRE- (Sodir et al., 2011)
Omomyc;CMVrtTA;RIP1-
Tag2
Amplification Mast cells [ MYC activation drives IL-1b and CCL5 Pancreatic cancer pIns-mycERTAM;RIP7- (Shchors et al., 2006;
expression, leading to an influx of mast cells bcl-xL Soucek et al., 2007)
in the pancreatic tumor
CD4+ T cells Y Regulates expression of CD47 and PDL1 T-ALL MYC T-ALL s.c. (Casey et al., 2016)
transplanted cell line, Em-
tTA/tet-O-MYC, LAP-tTA/
tet-O-MYC
Macrophages [ NK cells Y MYC drives expression of CCL9, which recruits NSCLC KrasLSL-G12D;Rosa26-LSL- (Kortlever et al., 2017)
macrophages, and IL-23, which limits NK MycERT2
recruitment
T, B cells Y MYC drives expression of IL-23, which excludes
T and B cells from the tumor
NOTCH Amplification Macrophages [ NOTCH activates CCL2 and IL-1b production by Breast cancer 4T1, MDA-MB-231 cell (Shen et al., 2017)
tumor cells thus increasing pro-tumoral monocytes lines and RBPJkIND-
and macrophages MMTV;MMTV-PyMT
T cells Y NOTCH represses CEBP/b leading to impaired Liver cancer Hydrodynamic tail-vein (Hoare et al., 2016)
clearance of senescent cells and subsequent injection of NRASG12V
liver tumor development
NRAS Mutation Neutrophils, monocytes, NRAS mutation induces SASP, thus recruiting Liver cancer Hydrodynamic tail-vein (Kang et al., 2011)
NK cells, macrophages, immune cells and CD4+ T cell-mediated injection of NrasG12V
DCs [ clearance of tumor cells. and NrasG12V/D38A
CD4+ T cells [ NRAS-induced senescent cells are cleared by
CD4+ T cells
p53 Mutation Myeloid cells [ Mutant p53 activates NFkB and thus drives CRC DSS-induced (Cooks et al., 2013)
cytokine production and inflammation-
associated tumor progression
Loss Neutrophils, macrophages [ Potentially via dysregulation of NFkB Lung cancer KrasLSL-G12D/+ and (Meylan et al., 2009)
KrasLSL-G12D/+;Trp53F/F
Immunity 48, March 20, 2018 405

Macrophages [ Loss of p53 leads to an impaired intestinal epithelial CRC Villin-creERT2;Trp53F/F (Schwitalla et al., 2013)
barrier, thus triggering intestinal microflora- and AOM-induced
mediated immune activation via NFkB.
Macrophages, monocytes, STAT3-mediated establishment of an PDAC Ptfa1-cre;KrasLSL-G12D/+, (Wörmann et al., 2016)
neutrophils [ immunosuppressive microenvironment Ptfa1-cre;KrasLSL-G12D/+;
p53F/F and Ptfa1-cre;
KrasLSL-G12D/+;p53R172H/+
Monocytes [ p53 transcriptionally regulates CXCL17, and loss Prostate cancer Pb-cre;Pten F/F;Trp53F/F (Bezzi et al., 2018)
of p53 leads to an increase of CXCL17, thus
recruiting monocytes to the tumor
(Continued on next page)
406 Immunity 48, March 20, 2018

Table 2. Continued
Genetic Consequence for
Gene aberration intratumoral immune cells Signaling involved Tumor type Tumor model Reference
PRKCI Amplification NK cells Y, CD11b+Gr1+ PRKCI activates YAP1, inducing TNFa to promote High-grade Pax8-cre;tetOLSL-PRKCI; (Sarkar et al., 2017)
cells [ an immunosuppressive microenvironment serous ovarian PtenF/F;Trp53F/F with
CD8+ T cellsY PRKCI amplification induces immunosuppressive carcinoma inducible loss of PRKCI
neutrophils, thus reducing CD8+ T cells and cell lines derived
from these tumors
PTEN Loss CD11b+Gr1+ cells [ PTEN loss activates NFkB and thereby expression PDAC p48-Cre; KrasLSL-G12D; (Ying et al., 2011)
of CXCL1, G-CSF, IL-23 PtenF/+
CD8+ T cells Y Loss of PTEN promotes resistance to T cell killing Melanoma Cell line inoculation (Peng et al., 2016)
by inhibiting autophagy models and Tyr:CreER;
PtenF/F;Braf V600E/+
RAS Mutation CD11b+Gr1+ cells [ Via GM-CSF production by tumor cells PDAC KrasG12D inoculation (Ancrile et al., 2007;
model Pylayeva-Gupta et al.,
2012)
RB Loss Macrophages Y NA SCLC Rb1F/F;Trp53F/F (Busch et al., 2016)
SMAD4 Loss Neutrophils [ SMAD4 loss increases YAP1-mediated CXCL5 Prostate cancer Pb-cre;PtenF/F;Trp53F/F (Wang et al., 2016)
expression, thus driving immunosuppressive
neutrophils.
CD8+ T cells, Tregs Y PRKCI amplification induces immunosuppressive
neutrophils, thus reducing CD8+ T cells and Tregs
ZBTB7a Loss Neutrophils [ p53 transcriptionally regulates SOX-9, and loss of Prostate cancer Pb-cre;Pten F/F;Trp53F/F (Bezzi et al., 2018)
p53 leads to an increase of SOX-9, which in turn
activates CXCL5, thus recruiting neutrophils to
the tumor
Abbreviations: NA, Not assessed. SASP, Senescence-associated secretory phenotype. CRC, Colorectal cancer. NSCLC, Non-small cell lung cancer. PDAC, Pancreatic ductal adenocarcinoma.
T-ALL, T cell acute lymphoblastic leukemia. SCLC, small cell lung carcinoma.
Listed here are the cancer cell-intrinsic genetic aberrations that result in a change in innate and adaptive immune contexture as demonstrated in genetically engineered mouse models.

Review
Immunity
Immunity

Review

MYC regulates many essential processes in the cell. In addition, suppression of interferons and pro-inflammatory signaling path-
recent studies revealed that it also has a strong hold on the ways (Layer et al., 2017). These studies show that MYC activa-
tumor immune landscape (Figure 1, Table 2). Using the RIP1- tion in tumors can control immune checkpoint molecules and
Tag2;TRE-Omomyc;CMV-rtTA pancreatic b-cell cancer mouse T cell influx, but the underlying mechanisms may differ between
model, in which treatment with doxycyclin induces expression tumor types.
of a dominant-negative MYC mutant, it was shown that inhibition Another mechanism by which MYC regulates the immune
of endogenous MYC in established islet tumors resulted in tumor phenotype of tumors was recently demonstrated in the
regression, which was accompanied by a marked decrease in KrasG12D-driven lung adenocarcinoma model. Here, conditional
infiltrating macrophages and neutrophils (Sodir et al., 2011). MYC amplification resulted in a rapid decrease of intratumoral B,
This study illustrates that although MYC is not an oncogenic T, and NK cells, and an increase in macrophages (Kortlever et al.,
driver in this tumor model, its endogenous expression is crucial 2017). Mechanistically, MYC amplification led to increased
for tumor progression and has a profound effect on the inflam- expression of IL-23 by cancer cells, which inhibited B, T, and
matory microenvironment. In another transgenic b-cell cancer NK cell recruitment, and increased expression of CCL9, which
mouse model carrying a switchable form of the MYC oncopro- recruited and activated macrophages in the tumor. These mac-
tein in the pancreas, forced expression of MYC in b-cells resulted rophages inhibited T cells, while also promoting angiogenesis.
in pancreatic cancer formation (Shchors et al., 2006). Impor- Interestingly, these tumors rapidly acquired dependency on
tantly, Myc activation stimulated production of the potent pro-in- MYC amplification, and MYC de-activation resulted in tumor
flammatory cytokines CCL5 and interleukin-1b (IL-1b) by b cells, regression in an NK cell-dependent fashion (Kortlever et al.,
which facilitated tumor angiogenesis and recruitment of pro- 2017). These findings suggest that targeting MYC in tumors
tumoral mast cells to the tumor (Shchors et al., 2006; Soucek would be an attractive therapeutic strategy to unleash anti-tumor
et al., 2007). These studies demonstrate that MYC can drive immunity. While MYC is as of yet not directly targetable, indirect
tumor progression at least in part through orchestrating pro- therapeutic strategies emerge. One such strategy targets the
tumoral inflammatory conditions. epigenetic modulators DNA methyl transferases (DNMTs) and
The effects of MYC signaling on the tumor microenvironment histone deacetylases (HDACs). Combined treatment of NSCLC
may not be limited to pancreatic cancer alone. In the Em-tTA- mouse models with DNMT and HDAC inhibitors reduced MYC
TRE-Myc mouse lymphoma model, inactivation of MYC in estab- expression, increased CCL5 levels, decreased macrophage
lished tumors resulted in a marked decrease in intratumoral influx, and increased cytotoxic T cell influx and inhibited tumor
macrophages (Yetil et al., 2015). It would be of interest to assess growth (Topper et al., 2017). This study demonstrates that indi-
whether the same MYC-controlled inflammatory mediators are rect targeting of MYC might prove therapeutically beneficial by
involved in lymphoma and pancreatic cancer. Interestingly, limiting tumor growth and reversing immune evasion. However,
upon additional loss of p19ARF, but not p53, MYC-dependent this study did not formally exclude a direct effect of the epige-
regulation of macrophage recruitment is not observed (Yetil netic modulators on the immune system.
et al., 2015), suggesting that the ability of MYC to control recruit- These studies show that in addition to the key role MYC has in
ment of immune cells to tumors can be counteracted by other tumor cell-intrinsic processes, this transcription factor can exert
aberrantly expressed genes. This is also illustrated by the obser- a wide variety of functions to modulate both the innate and the
vation that the spontaneous anti-tumor T and NK cell response in adaptive immune landscape of several tumor types. While
the Em-MYC lymphoma model could only be elicited when Bcl-2 MYC is not directly targetable, insights into these mechanisms
was overexpressed, but not when p53 was deleted (Schuster open up new ways to target MYC-regulated signaling.
et al., 2011). How p53 loss counteracts MYC activity in modu-
lating the tumor microenvironment however remains a subject Other Genetic Determinants of the Tumor Immune
of future research. Landscape
MYC can also control the immune landscape of tumors by The effect of oncogenes and TSGs on the tumor immune land-
regulating expression of immune checkpoint molecules. In the scape is not just limited to the abovementioned genes and path-
Em-tTA/tet-O-MYC lymphoma model and cell lines with switch- ways; several other genetic events and downstream immune
able MYC expression, MYC increased the expression of both effects have been described (Figure 1, Table 2). One example
PD-L1 and the ‘‘don’t eat me’’ receptor CD47 on cancer cells is the impact of the Ras oncogene on tumor-associated myeloid
by binding directly to their respective promoters (Casey et al., cells. Mutated Ras strongly induces expression of IL-6 and IL-8
2016). Exogenous overexpression of PD-L1 and CD47 on cancer in in vitro models (Ancrile et al., 2007; Sparmann and Bar-Sagi,
cells limited the CD4+ T cell and macrophage recruitment to the 2004). These Ras-controlled cytokines have been reported to
tumor. Moreover, MYC inactivation downregulated CD47 and facilitate myeloid cell infiltration and tumor progression (Ancrile
PD-L1 expression and induced tumor regression, while exoge- et al., 2007; Sparmann and Bar-Sagi, 2004). Furthermore,
nous overexpression of PD-L1 and CD47 in cancer cells KrasG12D-induced changes in cytokine expression resulted in
enhanced disease progression (Casey et al., 2016). Although accumulation of CD11b+Gr1+ immunosuppressive cells in a
not experimentally proven, this study suggests that MYC may variety of tumor models, including pancreatic and lung cancer
facilitate tumor immune escape by induction of immune check- (Ji et al., 2006; Pylayeva-Gupta et al., 2012; Wislez et al.,
points. Similarly, a MYC amplification-dependent T cell-poor 2006). Ablation of one of the KrasG12D-induced cytokines, GM-
environment has also been reported in human neuroblastomas, CSF, in tumor cells impaired immunosuppressive cells from
but in these tumors genomic amplification of N-MYC inversely entering pancreatic tumors and consequently resulted in an in-
correlated with PD-L1 expression, possibly due to MYC-induced crease in CD8+ T cells (Pylayeva-Gupta et al., 2012). These

Immunity 48, March 20, 2018 407


Immunity

Review

studies demonstrate the causal relationship between Ras onco- tions in the tumor. With the rapid developments in mouse
genic signaling pathways, immune-stimulatory transcription pro- model-generating techniques (Huijbers, 2017), future systematic
grams and immune landscape. Another study revealed a role for approaches in GEMMs may increasingly reveal causal geno-
adherence junction protein a-catenin in inflammatory signaling. type-immunophenotype relationships, and its impact on tumor
In the K14-Cre;a-cateninF/F mouse model for skin squamous progression.
cell carcinoma (SCC), loss of a-catenin activates NFkB and
its downstream inflammatory target genes, such as IL-1b and The Role of Oncogene-Induced Senescence in
IL-6, and stimulates SCC, thus again linking tumor-initiating Promoting an Inflammatory Tumor Microenvironment
oncogenic events with NF-kB-mediated immune signaling (Ko- A cancer-cell-intrinsic pathway in which many of the above-
bielak and Fuchs, 2006). Likewise, by comparing the Pdx1- mentioned cancer-driving genes are involved and that strongly
cre;KrasLSL-G12D and the Pdx1-cre;KrasLSL-G12D;Pten+/F mouse influences the intratumoral immune landscape is cellular senes-
models for pancreatic cancer, it was demonstrated that loss of cence. In a process called oncogene-induced senescence (OIS),
Pten resulted in increased activation of the NF-kB pathway, precancerous cells undergo cell-cycle arrest upon activation of
driving expression of several immune regulators by cancer cells, oncogenic signaling. Cellular senescence is a physiological pro-
such as G-CSF, IL-23 and CXCL1 (Ying et al., 2011). Pten loss gram that can be activated in response to cellular stress and ag-
and the downstream NF-kB activation not only accelerated tu- ing, leading to an essentially irreversible cell proliferation arrest
mor progression, but also influenced the frequency of intratu- (Muñoz-Espı́n and Serrano, 2014). Senescent cells can persist
moral neutrophils, monocytes, and Tregs (Ying et al., 2011). and actively secrete cytokines and other inflammatory and
Another study showed a profound role for the STK11/LKB1 growth-promoting factors, a process called the senescence-
tumor suppressor in NSCLC. Comparing KrasG12D/+ with associated secretory phenotype (SASP) (Pérez-Mancera et al.,
KrasG12D/+;Lbk1 / mice, it was found that loss of Lkb1 resulted 2014). Through their SASP, senescent cells can exert a signifi-
in increased IL-6 production, which resulted in higher intratu- cant, and sometimes opposing, impact on the immune land-
moral and systemic immunosuppressive neutrophil levels scape of the tumor. SASP can lead to immune-mediated
(Koyama et al., 2016). Indeed, blockade of IL-6 resulted in clearance of pre-malignant cells, or via stimulation of chronic
increased levels cytotoxic CD8+ T cells and tumor control inflammation promote tumor progression. Below we discuss
(Koyama et al., 2016). Although not all of these studies eluci- how oncogenes and TSGs, via SASP activation, shape the in-
dated the functional consequence of the altered immune land- flammatory microenvironment.
scape on tumor growth, they demonstrate that a wide variety Several oncogenes and TSGs have been linked with SASP
of cancer-driving mutations can dictate the composition of the activation (Figure 2). The p53 pathway plays an important role
tumor microenvironment. in the induction of OIS. This was demonstrated by the induction
Collectively, studies pertaining to cancer cell-intrinsic path- of senescence and tumor clearance upon doxycyclin-mediated
ways and immune contexture are gaining ground and have iden- activation of p53 in a HrasG12V;TRE.shp53 inoculation model
tified various cancer-driving genes that orchestrate diverse for liver cancer (Xue et al., 2007). Activation of p53 did not lead
immune landscapes in the tumor. Thus far, many of these studies to tumor cell death in a cell-autonomous manner, but rather neu-
have been relatively biased and focused on a single genetic trophils, macrophages and NK cells were recruited to these tu-
pathway in a single mouse tumor model. A more systematic mors by activated SASP and removed the senescent cells (Xue
assessment of immune cell populations in relation to tumor et al., 2007). Indeed, maintenance of WT p53 was a prerequisite
genotypes was recently performed in two studies. One com- of senescence induction, as also observed in other tumor
pared four independent lung cancer GEMMs: Ccsp-rtTA;TetO- models (Cooks et al., 2013; Pribluda et al., 2013). Because
EgfrL858R, Rb1F/F;Trp53F/F, KrasLSL-G12D/+ and KrasLSL-G12D/+; NF-kB is a key transcription factor in SASP activation (Chien
Trp53F/F models, representing molecularly distinct human et al., 2011), the regulation of NF-kB by the p53 pathway might
SCLC and NSCLC subtypes (Busch et al., 2016). This approach play an important role in SASP regulation. In colorectal tumor
revealed key differences in immune cell content between the models, Wnt signaling can also regulate SASP induction. Villin-
different tumor genotypes, such as that EgfrL858R-driven tumors creERT2;CKIaF/F mice, which display hyper-activated Wnt
showed lower frequencies and activation of CD8+ T cells signaling due to loss of CKIa, exhibit growth arrest of colorectal
compared to Kras-driven tumors, whereas NK cells in Kras- tumors and induction of senescence, paired with an inflamma-
driven tumors, but not EGFR mutants, show downregula- tory response (Pribluda et al., 2013). SASP is maintained upon
tion of activation markers (Busch et al., 2016). A second additional p53 deletion in this model, however, it dissociates
study compared the Pb-cre;PtenF/F;Zbtb7aF/F, Pb-cre;PtenF/F; from growth arrest while the inflammatory response continues,
Trp53F/F and Pb-cre;PtenF/F;PmlF/F prostate cancer models resulting in inflammation-accelerated tumorigenesis (Pribluda
and observed profound differences in composition of the tumor et al., 2013). These findings illustrate that depending on the
microenvironment (Bezzi et al., 2018). Mechanistic studies re- genetic makeup of cancer cells, the senescence-associated in-
vealed distinct chemokine production by tumors controlled by flammatory response can result in two opposing outcomes: tu-
loss of Zbtb7a, p53, or Pml and blockade of the respective mor inhibition or tumor promotion. In addition to p53 and Wnt,
signaling pathways impaired innate immune cell recruitment mTOR signaling was shown to induce SASP in CRC and prostate
and tumor progression. These studies demonstrate the powerful cancer cells in vitro (Laberge et al., 2015). mTOR inhibition by ra-
potential of GEMMs in identifying the complex mechanisms that pamycin decreased mTOR-induced SASP and decreased influx
control the tumor microenvironment and potential for immuno- of macrophages, T, B, and NK cells into inoculated NrasG12V
modulatory therapeutic intervention based on genetic aberra- mutant liver tumors (Herranz et al., 2015). These studies suggest

408 Immunity 48, March 20, 2018


Immunity

Review
Figure 2. Relationship between Genetic
A Rasmut B Legend
Events in Cancer Cells, the Dynamic
Ras mut
p53WT MYC Aspects of SASP and the Immune System
mTOR p53LOF
Wntactive NK cell (A) Oncogene-induced senescence (OIS), in
combination with WT p53, activated MYC, low
CD8 T cell
+
Notch signaling, active Wnt signaling, activated
NF B RAS, or active mTOR signaling induces a senes-
STAT3
Macrophage
cence-associated secretory phenotype (SASP)
Notchlow Senescence-associated that leads to the recruitment and activation of
Notchhigh
Non-senescent secretory phenotype Non-senescent
malignant cell malignant cell macrophages, neutrophils, NK cells, and CD8+
Neutrophil
T cells that clear senescent cells and thus limit
Chronic inflammation
Immune-mediated & immunosuppresion
tumorigenesis.
clearance of (B) Loss or loss-of-function mutations in p53, or
senescent cells
activated RAS, Notch, or mTOR signaling can
lead to an alternative SASP that also attributes to
a chronic inflammatory state that establishes an
immunosuppressive tumor microenvironment.
Immunosuppressive macrophages and neutro-
Senescent cell
phils limit NK and CD8+ T cell-mediated anti-tu-
Tumor mor response and thus promote tumorigenesis.
NF-kB and STAT3 signaling in senescent cells is
key in SASP induction.

that targeted therapies, such as rapamycin, may reduce tumor- cells can actively counteract SASP by producing IL-1 receptor
induced inflammation, but potentially also reduce senescent antagonist (Di Mitri et al., 2014). Additionally, senescence pro-
tumor cell clearance by infiltrating immune cells, thus demon- grams in tumor-associated stromal cells also impact tumorigen-
strating the complexity of targeting SASP. Nonetheless, these esis through modulation of immune responses. In a carbon
studies reveal the essential role of oncogenes and TSGs in tetrachloride (CCl4)-induced liver fibrosis model, p53 activity in
SASP induction and the potential of targeting these genes to hepatic stellate cells (HSCs) limits fibrosis and cirrhosis, and
revert tumor-promoting SASP. reduced liver tumorigenesis in mice treated with CCl4 and
The composition of SASP mediators secreted by senescence diethylnitrosamine (DEN) (Lujambio et al., 2013). Here, wild-
cells is dynamic and experimental evidence points toward type p53 cooperated with NF-kB to induce senescence and
NOTCH1 as one of the master regulators controlling this SASP SASP in HSCs, which induced a tumor-inhibiting phenotype in
diversity. In NrasG12V mutant tumor models, NrasG12V-induced macrophages. Loss of p53 in stromal HSCs changed their secre-
senescence was accompanied by fluctuations in endogenous tome, induced the polarization of macrophages toward a tumor-
Notch expression levels (Hoare et al., 2016). Ectopic expression promoting phenotype and accelerated inflammation-induced
of active Notch in an NrasG12V-dependent oncogene-induced hepatocellular carcinoma (Lujambio et al., 2013), indicating
senescence liver model increased cancer progression in a that also stromal cell-intrinsic p53 controls tumorigenesis via
non-cell-autonomous fashion (Hoare et al., 2016). In this model, modulation of the immune system.
Notch levels determined the composition of the SASP and sub- Collectively, depending on the tumor type and oncogenic wir-
sequent immune function. Notch inhibited lymphocyte-mediated ing, the activated SASP-related genes and downstream inflam-
clearance of senescent cells through repression of C/EBPb. matory profile may differ, resulting in a wide spectrum of immune
Reversely, inhibition of Notch during senescence led to an in- responses that range from tumor-promoting chronic inflamma-
crease of lymphocyte-mediated senescent cell clearance (Hoare tory responses to immune-mediated clearance of cancer cells
et al., 2016). This Notch-dependent cytokine production and (Figure 2). Deeper mechanistic insights into the causal relation-
shaping of the immune phenotype of tumors was also demon- ship between genetic events in cancer cells and the dynamic
strated in breast cancer, where tumor-intrinsic Notch signaling aspects of SASP may open new avenues for therapeutic inter-
increased monocyte and macrophage accumulation by in- vention. Indeed, this is exemplified by a study showing that the
creasing expression of IL-1b and CCL2 (Shen et al., 2017). These efficacy of docetaxel could be enhanced by pharmacologically
studies demonstrate that immune cell influx can be strongly targeting Pten-loss-induced SASP in a transgenic prostate tu-
influenced by SASP, but also that the activity of cancer cell- mor model (Toso et al., 2014). Important to note however, is
intrinsic genes play important roles in determining the spectrum that senescent cells are not the only cells actively secreting in-
of inflammatory mediators produced within the tumor. Indeed, in flammatory mediators in the tumor, and the cytokine milieu
the Ptf1a-cre;KrasLSL-G12D/+ mouse model for pancreatic cancer, and its net effect on the immune landscape is not only deter-
genetic deletion of RelA, the gene that encodes the NF-kB sub- mined by SASP. Therefore, it is of key importance to delineate
unit p65, abrogated senescence and SASP, thus enhancing pro- how the tumor-promoting aspects of SASP can be reverted,
gression of pancreatic tumors (Lesina et al., 2016). While while enhancing the tumor-limiting aspects.
reducing SASP, RelA deletion led to a marked increase in
immunosuppressive cells and decreased T cell activation in Mechanisms of Cancer-Cell-Intrinsic Regulation of
the pancreata of these mice (Lesina et al., 2016). Therefore, Parameters of the Cancer Immunity Cycle and Immune
in these tumors, the cancer-immune cell crosstalk is not limited Checkpoint Blockade Response
to SASP. As discussed above, the mutational load of tumors is one of
The infiltrating immune cells can also impact senescence it- the determinants linked with responsiveness to immune check-
self. In Pten-induced senescent prostate tumors, CD11b+Gr-1+ point inhibition. The expectation is that many other parameters,

Immunity 48, March 20, 2018 409


Immunity

Review

including the activation of certain oncogenes or inactivation of prove beneficial to alleviate T cell suppression. Collectively,
TSGs, are associated with therapeutic benefit as well, and that these studies show that oncogenic pathway activation can
they may differ per tumor (sub)type. As of yet, preclinical studies significantly impact on parameters of the cancer-immunity cycle.
focused on unlocking the relationship between tumor genetics However, the functional consequences of these genetic changes
and response to immunotherapy are still relatively limited, how- on immunotherapy response have not been addressed in these
ever, the concept is emerging that genetic events in cancer cells studies. Focal Adhesion Kinase (FAK) activity in cancer cells has
dictate various aspects of the tumor-immunity cycle (Chen and also been identified as an important regulator of immunosup-
Mellman, 2013), such as activation of immunosuppressive pression in the tumor microenvironment, and its impact on
myeloid cells, induction of immune checkpoint molecule expres- immunotherapy efficacy has been addressed experimentally.
sion, regulation of DC activation and T cell priming, and induction FAK amplification was observed in the p48-Cre;KrasLSL-G12D;
of tumor resistance to T cell attack. Trp53F/+ model for PDAC, and therapeutic targeting of FAK
One such genetic event is mutation in the serine/threonine- improved survival by alleviating the immunosuppressive micro-
protein kinase ATR. ATR is a DNA damage sensor and is environment, mainly by reducing macrophages, monocytes,
frequently mutated in melanoma. It has been reported to influ- and neutrophils in the tumor (Jiang et al., 2016). This held true
ence important parameters of immunotherapy response, such for cancer-cell-specific ablation of FAK, indicating that immune
as intratumoral T cell influx and expression of immune check- cell changes occur via FAK targeting in cancer cells. Importantly,
points. Transgenic expression of an ATR LOF mutant in the inhibition of FAK synergized with anti-CTLA-4/anti-PD-1 combi-
Tyr::CreERT2; BrafV600E;PtenF/F model for melanoma diminished nation immunotherapy (Jiang et al., 2016), indicating that inter-
T cell influx in the tumor, while increasing B cells and macro- ference with this cancer cell-intrinsic signaling pathway renders
phages (Chen et al., 2017). This was associated with an increase tumors sensitive to immunotherapy.
in expression of Arginase 1, CD206, and PD-L1 in the tumor, DC activation and T cell priming can also be influenced by
suggesting a more T cell suppressed environment. Cyclin- cancer cell-intrinsic signaling pathways. Using the BrafV600E;
dependent kinases (CDKs)—essential regulators of the cell Pten–/–;CAT-STA mouse model for melanoma, which expresses
cycle—have also been shown to be involved in immune check- constitutively active b-catenin, it was revealed that b-catenin
point regulation. In medulloblastoma (MB) cell line inoculation signaling prevented expression of CCL4 by cancer cells, result-
models, the anti-tumor function of CD4+ T cells depends on ing in suppression of recruitment of CD103+ DCs and impaired
disruption of CDK5 in MB cells (Dorand et al., 2016). In this priming and intratumoral accumulation of T cells (Spranger
model, CDK5 is required for PD-L1 expression by MB cells, as et al., 2015). As a consequence, b-catenin-active tumors failed
CDK5 is a repressor of IRF2 and IRF2BP2, that both regulate to respond to anti-CTLA-4/anti-PD-1 treatment. In line with these
IFN-g-mediated PD-L1 expression (Dorand et al., 2016). Addi- data, active WNT/b-catenin signaling in human metastatic mela-
tionally, it was recently shown that the activating RasG12V muta- nomas correlated with absence of a T cell gene expression
tion can cause stabilization of PD-L1 mRNA via activation of signature (Spranger et al., 2015). This study highlights the impor-
MEK (Coelho et al., 2017). However, the functional relevance of tance of cancer cell-intrinsic WNT/b-catenin signaling in immune
these changes for immunotherapy and disease progression in evasion of tumors, and suggests that targeting the WNT pathway
relation to ATR, CDK5, and RAS remains unaddressed in these may improve the therapeutic benefit of immune checkpoint inhi-
studies. bition in tumors with active b-catenin signaling.
Another mechanism by which tumor cells may regulate immu- Some oncogenes and TSGs have been demonstrated to regu-
notherapy response is via establishment of an immunosuppres- late immune checkpoint molecule expression in a cell-autono-
sive microenvironment. Overexpression of PRCKI, a protein mous fashion, and thus influence response to immunotherapy.
kinase, is frequently observed in a variety of cancer types, In EGFR-driven lung cancer mouse models, EGFR mutation
including high-grade serous ovarian carcinoma (Sarkar et al., caused rapid induction of an immunosuppressive tumor micro-
2017). Upon conditional overexpression of PRKCI in the Pax8- environment (Akbay et al., 2013). The EGFR mutant lung tumors
rtta;TetO-Cre;Trp53F/F;PtenF/F mouse model for ovarian cancer, displayed increased expression of immune checkpoint mole-
tumors upregulated TNF-a, as a result of which tumors were cules such as PD-1 and PD-L1, which led to an increased sensi-
strongly infiltrated by immunosuppressive neutrophils, thus tivity to anti-PD-1 monotherapy in these tumor-bearing mice. In
decreasing CD8+ T cell influx (Sarkar et al., 2017). This TNF-a- line with these pre-clinical findings, EGFR pathway activating
mediated neutrophil recruitment was dependent on PRKCI- mutations in human lung tumors, and not the other prevalent
induced YAP1—a key transcriptional regulator and onco- driver mutation KRASG12V, correlated with PD-L1 expression
gene—signaling in cancer cells (Sarkar et al., 2017). Likewise, (Akbay et al., 2013). Intriguingly, another study reported KRAS
by comparing Pb-cre4;PtenF/F with Pb-cre4;PtenF/F;Smad4F/F mutant lung tumors in patients treated with anti-PD-1 to have
prostate cancer mouse models, a strong YAP1-dependent influx higher PD-L1 levels relative to EGFR mutated tumors (Garon
of neutrophils was observed upon cancer cell-intrinsic Smad4 et al., 2015), potentially mediated by KRAS-induced stabilization
loss (Wang et al., 2016). Here, Smad4 loss caused YAP1- of PD-L1 (Coelho et al., 2017). The different levels of PD-L1 regu-
mediated upregulation of CXCL5 in tumor cells. This in turn re- lation by mutated oncogenes and the underlying mechanisms
cruited CXCR2+ neutrophils, which suppressed the CD8+ T cell will therefore be an important topic of future research.
response to the tumor (Wang et al., 2016). These studies show Similarly, PTEN status is implicated in immunotherapy
that Smad4 and PRCKI both function as inducers of immunosup- response due to its ability to render cancer cells resistant to
pression via cancer cell-intrinsic YAP signaling and that YAP in- T cell attack. In a cohort of melanoma patients, PTEN loss corre-
hibitors—which are currently in preclinical development—may lated with low TIL influx and poor response to anti-PD-1 therapy

410 Immunity 48, March 20, 2018


Immunity

Review
Figure 3. How to Exploit the Genetic
A B Makeup of Individual Tumors to Allow for
Patient-Specific Immune-Based
Therapeutic Interventions
Maximizing therapeutic efficacy by rational se-
lection of targeted drugs and immunomodulatory
compounds based on the genetics of the tumor.
Examples depicted here are mainly based on pre-
clinical intervention studies, with therapeutic mo-
dalities highlighted in red. For every example a
mouse or human symbol is used to depict what is
based on clinical or pre-clinical evidence.
(A) In breast cancer, CDK4/6 inhibition increases
D
antigen presentation, interferon signaling, and
CD8+ T cell levels, while decreasing Tregs in the
C tumor. Combined with anti-PD-L1 treatment, this
leads to a marked tumor regression (Goel et al.,
2017).
(B) In EGFR mutant lung cancer, PD-L1 has been
described to be upregulated, increasing the
sensitivity to anti-PD-L1 therapy (Akbay et al.,
2013). KRAS mutation in lung cancer can also
drive PD-L1 expression, to a higher extent than
EGFR mutation (Garon et al., 2015). In MYC-driven
lung tumors, combined inhibitors against HDAC
E and DNMT both target MYC and CD8+ T cells, thus
limiting tumor growth (Topper et al., 2017).
(C) Pancreatic tumors with FAK amplification
show an accumulation of immunosuppressive
cells in the tumor. FAK1/2 inhibitors alleviate this,
and combined with anti-PD-1 and anti-CTLA-4
treatment limit tumor progression (Jiang et al.,
2016). Pancreatic tumors with p53 loss or muta-
tion establish an immunosuppressive microenvi-
ronment by JAK-STAT signaling. Targeting JAK2
in combination with gemcitabine reduces tumor
burden (Wörmann et al., 2016).
(D) In melanoma, ATR loss-of-function mutation
increases PD-L1 and thereby potentially sensi-
tizes these tumors to anti-PD-L1 treatment. In
PTEN null melanomas, the resulting activated AKT signaling can be reduced by PI3Kb inhibitors, which in combination with anti-PD-1 limits tumor growth (Peng
V600E
et al., 2016). Combining MEK and BRAF inhibitors in BRAF mutant melanoma also synergize with anti-PD-1 treatment (Hu-Lieskovan et al., 2015).
(E) In prostate tumors with loss of SMAD4, YAP1-mediated immunosuppressive neutrophil recruitment can be counteracted by YAP1 inhibitors or anti-CXCR2
treatment (Wang et al., 2016).

(Peng et al., 2016). Using xenograft mouse models for mela- those with mutations in DNA repair machinery, or mutations
noma, it was shown that PTEN loss in cancer cells reduced can be engineered in a tissue-specific manner. This would allow
T cell influx, and resulted in reduced autophagy, leading to resis- for physiological modeling and therefore correct assessment of
tance to T cell-mediated killing (Peng et al., 2016). Treating PTEN pre-clinical immunotherapeutic strategies in an immunocompe-
null tumors with a PI3Kb inhibitor, thus reducing the dysregu- tent setting.
lated AKT activity in these tumors, improved response to anti-
PD-1 therapy, highlighting a potential therapeutic approach for Targeting Genetic Pathways to Unleash Anti-Tumor
PTEN null melanoma in controlling resistance to anti-PD-1 Immunity
therapy. One major theme that emerges from the aforementioned studies
Altogether, these studies show that aberrant signaling path- is that many targeted therapies, specific for hyperactive
ways in cancer cells can impact the anti-cancer immune signaling pathways, are likely to also exert a major impact on
response and the response to immune checkpoint inhibition the immune contexture of tumors. Most targeted drugs initially
(Figure 3). One aspect that needs to be taken into account induce very strong anti-cancer effects in patients, however, the
when using GEMMs to model human cancers with high muta- rate of durable clinical responses is disappointingly low (Groe-
tional load, is that the mutational load in transgenic mice may nendijk and Bernards, 2014). Given the previously unrecognized
not correspond to that of the human tumors, due to the strong impact of these targeted drugs on the immune landscape of tu-
driver mutations engineered in these mice. This could be over- mors, the question arises whether we can rationally induce a
come by for example exposing early melanoma lesions to UV favorable immune environment in tumors or even sensitize tu-
irradiation, or early lung lesions to carcinogens. The drawback mors to immunomodulatory drugs by selective usage of targeted
however, is that this may not result in clonal antigens and the therapy. In this regard, we can learn from the growing number of
mutational spectrum may be highly variable from one mouse to pre-clinical studies that have addressed the impact of targeted
the next. Alternatively, transgenic models that are prone to drugs on the immune microenvironment of tumors and their
generate high mutational load tumors can be used, such as response to immunotherapy. For example, as described above,

Immunity 48, March 20, 2018 411


Immunity

Review

BRAF mutant thyroid tumors are characterized by infiltration of inhibition enhanced the efficacy of adoptive cell transfer and im-
immunosuppressive cells (Charoentong et al., 2017), raising mune checkpoint therapies by direct targeting of immunosup-
the question of whether inhibition of mutant BRAF in thyroid can- pressive neutrophils that express the cMET receptor (Glodde
cer would induce a more favorable immune contexture. Indeed, et al., 2017). However, targeting cMET-expressing neutrophils
combined targeting of BRAFV600E and SRC increased influx of in another study promotes tumor progression (Finisguerra
CD8+ T cells, B cells, and macrophages and reduced tumor et al., 2015), highlighting the complex model-dependent and
growth in an orthotopic inoculation model for anaplastic thyroid dual role of neutrophils in cancer biology (Coffelt et al., 2016).
cancer (Vanden Borre et al., 2014). Also in patients with Likewise, it has been reported that the depletion of immunosup-
BRAFV600E mutated metastatic melanoma, BRAF inhibition with pressive CD11b+Gr1+ cells as a bystander effect of other
vemurafenib enhanced melanoma antigen presentation by can- targeted therapies, for example by ITK/BTK-inhibitor ibrutinib,
cer cells, increased cytotoxic T cell influx, and decreased immu- benefits the response to immunotherapies in cell line inoculation
nosuppression (Frederick et al., 2013). This is in line with findings models for breast cancer and melanoma (Sagiv-Barfi et al., 2015;
in BRAFV600E melanoma mouse models in which BRAF inhibition Stiff et al., 2016). Ibrutinib can also reprogram macrophages,
improved adoptive T cell therapy (Koya et al., 2012) and BRAF relieve immunosuppression, and facilitate CD8+ cytotoxicity in
inhibition combined with MEK inhibition synergized with anti- PDAC-bearing mice (Gunderson et al., 2016). These studies
PD-1 treatment (Hu-Lieskovan et al., 2015). These studies highlight that targeted drugs can impact the immune contexture
indicate that therapeutic targeting of cancer cell-intrinsic of tumors via their working mechanism on cancer cells, which
oncogenic driver mutations can be exploited to induce a favor- indirectly changes the immune landscape, and via their direct
able immune environment and thus sensitize tumors to cancer effect on immune cells. Insights into the complexity of the com-
immunotherapy. bined effect of these targeted drugs on the cancer cells and tu-
Other targeted therapies have also been reported to exert mor microenvironment will help us to maximize the therapeutic
strong effects on the cancer-immune cell crosstalk. For benefit of targeted drugs in combination with immunomodula-
example, CDK4/6 inhibitors were originally designed to selec- tory strategies (Figure 3).
tively inhibit cell-cycle progression, but emerging experimental
evidence reveals that part of the therapeutic benefit of these in- Conclusions and Future Directions
hibitors lies in their anti-tumor immunity promoting capacity. In From the studies discussed in this review it has become clear
the MMTV-rtTA/tetO-HER2 mouse model for breast cancer, that activation of oncogenes or loss of TSGs not only exert an
treatment with the CDK4/6 inhibitor abemaciclib leads to tumor intrinsic influence on the fate of cancer cells, but can have pro-
regression by inducing anti-tumor immunity (Goel et al., 2017). found effects on tumor-host interactions. Commonly mutated
In vitro studies revealed that CDK4/6 inhibition increased antigen genes that lie at the basis of tumorigenesis can actively partici-
presentation and production of type III interferons by cancer pate in recruitment, activation, or dampening of the immune sys-
cells, which induced CD8+ T cell proliferation and activation tem. This could in part explain the heterogeneity between and
(Goel et al., 2017). Simultaneously, CDK4/6 inhibition reduced within tumor types in immune infiltration and activation. From a
systemic and intra-tumoral regulatory T cell numbers, which clinical perspective, these insights will help identify patients
occurred independent of the presence of a tumor. Both the effect that would or would not benefit from immunomodulation. More-
of the CDK4/6 inhibitor on antigen presentation by cancer cells over, identifying the mechanisms underlying the causal relation-
and the impact on regulatory T cells was dependent on inhibition ship between the genetic makeup of tumors and their immune
of the RB-E2F-DNMT1 axis (Goel et al., 2017). Importantly, by landscape may identify novel targets for anti-cancer immuno-
modulating the immune microenvironment, anti-CDK4/6 treat- modulatory therapies. The studies presented here likely only
ment improved response to anti-PDL1 in MMTV-rtTA/tetO- reveal the tip of the iceberg. Most studies focus on one particular
HER2 mice (Goel et al., 2017). Also, in an in vitro small molecule oncogene or TSG, and the majority of research is concentrated
screen, CDK4/6 inhibitors were identified to directly enhance on the primary tumor. This leaves the effect on the systemic im-
T cell activity. Mechanistically, CDK4/6 inhibition resulted in mune milieu and metastasis largely unaddressed. With increas-
de-repression of NFAT activity in T cells, resulting in increased ingly sophisticated methodologies to generate mouse models
T cell accumulation in lung tumors of KrasLSL-G12D;Trp53F/F that closely mimic the genetics and biology of human cancer
mice, which synergized with immune checkpoint inhibition and approaches to analyze tumors in depth, it will be possible
(Deng et al., 2018). These two studies illustrate that CDK4/6 to screen for a multitude of genetic and epigenetic alterations
inhibitors, which were originally developed to induce cell-cycle and their effect on the immune system. In vivo genetic manipula-
arrest in cancer cells, work in part by counteracting tumor im- tion will be key to delineate the spatiotemporal regulation of the
mune evasion. This is a result of combined targeting of cancer tumor immune landscape, both in the primary as well as the met-
cell-intrinsic pathways, changing parameters of the cancer-im- astatic lesion. This knowledge will help maximize the potential of
munity cycle, and direct targeting of T cells. immunomodulatory therapeutics for cancer patients and provide
Targeted therapies have also been reported to affect the abun- rationale for personalized combination therapies based on the
dance and function of myeloid cells in tumor-bearing hosts, genetic profile of tumors.
since the signaling pathways targeted by these drugs also play
functional roles in the immune system (Muñoz-Fontela et al.,
ACKNOWLEDGMENTS
2016). For example, neutrophils in the Hgf-Cdk4R24C model for
melanoma and cell line inoculation models impair the anti-tumor We apologize to those researchers whose original work could not be cited due
CD8+ T cell response (Glodde et al., 2017). In this study, cMET to space restrictions. We would like to thank Hannah Garner for insightful input

412 Immunity 48, March 20, 2018


Immunity

Review
during the writing process. Research in the De Visser laboratory is funded by Chen, D.S., and Mellman, I. (2013). Oncology meets immunology: the cancer-
European Research Council Consolidator award (InflaMet 615300), the Dutch immunity cycle. Immunity 39, 1–10.
Cancer Society (KWF10083; KWF10623), and the Beug Foundation for Metas-
tasis Research. K.E.d.V. is an EMBO Young Investigator. Chen, Z., Chen, X., Zhou, E., Chen, G., Qian, K., Wu, X., Miao, X., and Tang, Z.
(2014). Intratumoral CD8+ cytotoxic lymphocyte is a favorable prognostic
marker in node-negative breast cancer. PLoS ONE 9, e95475.
REFERENCES
Chen, C.F., Ruiz-Vega, R., Vasudeva, P., Espitia, F., Krasieva, T.B., de Fer-
audy, S., Tromberg, B.J., Huang, S., Garner, C.P., Wu, J., et al. (2017). ATR
Adams, J.M., Harris, A.W., Pinkert, C.A., Corcoran, L.M., Alexander, W.S., Mutations Promote the Growth of Melanoma Tumors by Modulating the
Cory, S., Palmiter, R.D., and Brinster, R.L. (1985). The c-myc oncogene driven Immune Microenvironment. Cell Rep. 18, 2331–2342.
by immunoglobulin enhancers induces lymphoid malignancy in transgenic
mice. Nature 318, 533–538. Chien, Y., Scuoppo, C., Wang, X., Fang, X., Balgley, B., Bolden, J.E., Pre-
msrirut, P., Luo, W., Chicas, A., Lee, C.S., et al. (2011). Control of the senes-
Akbay, E.A., Koyama, S., Carretero, J., Altabef, A., Tchaicha, J.H., Christen- cence-associated secretory phenotype by NF-kB promotes senescence and
sen, C.L., Mikse, O.R., Cherniack, A.D., Beauchamp, E.M., Pugh, T.J., et al. enhances chemosensitivity. Genes Dev. 25, 2125–2136.
(2013). Activation of the PD-1 pathway contributes to immune escape in
EGFR-driven lung tumors. Cancer Discov. 3, 1355–1363. Coelho, M.A., de Carné Trécesson, S., Rana, S., Zecchin, D., Moore, C., Mo-
lina-Arcas, M., East, P., Spencer-Dene, B., Nye, E., Barnouin, K., et al. (2017).
Ancrile, B., Lim, K.H., and Counter, C.M. (2007). Oncogenic Ras-induced Oncogenic RAS Signaling Promotes Tumor Immunoresistance by Stabilizing
secretion of IL6 is required for tumorigenesis. Genes Dev. 21, 1714–1719. PD-L1 mRNA. Immunity 47, 1083–1099.e6.

Bailey, P., Chang, D.K., Nones, K., Johns, A.L., Patch, A.M., Gingras, M.C., Coffelt, S.B., Wellenstein, M.D., and de Visser, K.E. (2016). Neutrophils in can-
Miller, D.K., Christ, A.N., Bruxner, T.J., Quinn, M.C., et al.; Australian Pancre- cer: neutral no more. Nat. Rev. Cancer 16, 431–446.
atic Cancer Genome Initiative (2016). Genomic analyses identify molecular
subtypes of pancreatic cancer. Nature 531, 47–52. Coley, W.B. (1893). The treatment of malignant tumors by repeated inocula-
tions of erysipelas: with a report of ten original cases. Am. J. Med. Sci. 105,
Balkwill, F., and Mantovani, A. (2001). Inflammation and cancer: back to 487–511.
Virchow? Lancet 357, 539–545.
Cooks, T., Pateras, I.S., Tarcic, O., Solomon, H., Schetter, A.J., Wilder, S., Loz-
Balli, D., Rech, A.J., Stanger, B.Z., and Vonderheide, R.H. (2017). Immune ano, G., Pikarsky, E., Forshew, T., Rosenfeld, N., et al. (2013). Mutant p53
Cytolytic Activity Stratifies Molecular Subsets of Human Pancreatic Cancer. prolongs NF-kB activation and promotes chronic inflammation and inflamma-
Clin. Cancer Res. 23, 3129–3138. tion-associated colorectal cancer. Cancer Cell 23, 634–646.

Becht, E., de Reyniès, A., Giraldo, N.A., Pilati, C., Buttard, B., Lacroix, L., Cooks, T., Harris, C.C., and Oren, M. (2014). Caught in the cross fire: p53 in
Selves, J., Sautès-Fridman, C., Laurent-Puig, P., and Fridman, W.H. (2016). inflammation. Carcinogenesis 35, 1680–1690.
Immune and Stromal Classification of Colorectal Cancer Is Associated with
Molecular Subtypes and Relevant for Precision Immunotherapy. Clin. Cancer Debebe, A., Medina, V., Chen, C.Y., Mahajan, I.M., Jia, C., Fu, D., He, L., Zeng,
Res. 22, 4057–4066. N., Stiles, B.W., Chen, C.L., et al. (2017). Wnt/b-catenin activation and macro-
phage induction during liver cancer development following steatosis. Onco-
Beroukhim, R., Mermel, C.H., Porter, D., Wei, G., Raychaudhuri, S., Donovan, gene 36, 6020–6029.
J., Barretina, J., Boehm, J.S., Dobson, J., Urashima, M., et al. (2010). The land-
scape of somatic copy-number alteration across human cancers. Nature 463, Decker, T., Fischer, G., Bu€cke, W., Bu €cke, P., Stotz, F., Gru€neberger, A.,
899–905. Gropp-Meier, M., Wiedemann, G., Pfeiffer, C., Peschel, C., and Götze, K.
(2012). Increased number of regulatory T cells (T-regs) in the peripheral blood
Bezzi, M., Seitzer, N., Ishikawa, T., Reschke, M., Chen, M., Wang, G., Mitchell, of patients with Her-2/neu-positive early breast cancer. J. Cancer Res. Clin.
C., Ng, C., Katon, J., Lunardi, A., et al. (2018). Diverse genetic-driven immune Oncol. 138, 1945–1950.
landscapes dictate tumor progression through distinct mechanisms. Nat.
Med. 24, 165–175. Deng, J., Wang, E.S., Jenkins, R.W., Li, S., Dries, R., Yates, K., Chhabra, S.,
Huang, W., Liu, H., Aref, A.R., et al. (2018). CDK4/6 Inhibition Augments Anti-
Blaisdell, A., Crequer, A., Columbus, D., Daikoku, T., Mittal, K., Dey, S.K., and tumor Immunity by Enhancing T-cell Activation. Cancer Discov. 8, 216–233.
Erlebacher, A. (2015). Neutrophils Oppose Uterine Epithelial Carcinogenesis
via Debridement of Hypoxic Tumor Cells. Cancer Cell 28, 785–799. Di Mitri, D., Toso, A., Chen, J.J., Sarti, M., Pinton, S., Jost, T.R., D’Antuono, R.,
Montani, E., Garcia-Escudero, R., Guccini, I., et al. (2014). Tumour-infiltrating
Boveri, T. (1914). Zur Frage der Entstehung Maligner Tumoren. Gustav Fischer Gr-1+ myeloid cells antagonize senescence in cancer. Nature 515, 134–137.
1914, 1–64.
Diakos, C.I., Charles, K.A., McMillan, D.C., and Clarke, S.J. (2014). Cancer-
Brandetti, E., Veneziani, I., Melaiu, O., Pezzolo, A., Castellano, A., Boldrini, R., related inflammation and treatment effectiveness. Lancet Oncol. 15,
Ferretti, E., Fruci, D., Moretta, L., Pistoia, V., et al. (2017). MYCN is an immu- e493–e503.
nosuppressive oncogene dampening the expression of ligands for NK-cell-
activating receptors in human high-risk neuroblastoma. OncoImmunology 6, Donehower, L.A., Harvey, M., Slagle, B.L., McArthur, M.J., Montgomery, C.A.,
e1316439. Jr., Butel, J.S., and Bradley, A. (1992). Mice deficient for p53 are developmen-
tally normal but susceptible to spontaneous tumours. Nature 356, 215–221.
Busch, S.E., Hanke, M.L., Kargl, J., Metz, H.E., MacPherson, D., and
Houghton, A.M. (2016). Lung Cancer Subtypes Generate Unique Immune Dorand, R.D., Nthale, J., Myers, J.T., Barkauskas, D.S., Avril, S., Chirieleison,
Responses. J. Immunol. 197, 4493–4503. S.M., Pareek, T.K., Abbott, D.W., Stearns, D.S., Letterio, J.J., et al. (2016).
Cdk5 disruption attenuates tumor PD-L1 expression and promotes antitumor
Casey, S.C., Tong, L., Li, Y., Do, R., Walz, S., Fitzgerald, K.N., Gouw, A.M., immunity. Science 353, 399–403.
€tgemann, I., Eilers, M., and Felsher, D.W. (2016). MYC regulates
Baylot, V., Gu
the antitumor immune response through CD47 and PD-L1. Science 352, Doucette, T., Rao, G., Rao, A., Shen, L., Aldape, K., Wei, J., Dziurzynski, K.,
227–231. Gilbert, M., and Heimberger, A.B. (2013). Immune heterogeneity of glioblas-
toma subtypes: extrapolation from the cancer genome atlas. Cancer Immunol.
Cha, Y.J., Kim, H.R., Lee, C.Y., Cho, B.C., and Shim, H.S. (2016). Clinicopath- Res. 1, 112–122.
ological and prognostic significance of programmed cell death ligand-1
expression in lung adenocarcinoma and its relationship with p53 status. Duesberg, P.H., and Vogt, P.K. (1970). Differences between the ribonucleic
Lung Cancer 97, 73–80. acids of transforming and nontransforming avian tumor viruses. Proc. Natl.
Acad. Sci. USA 67, 1673–1680.
Charoentong, P., Finotello, F., Angelova, M., Mayer, C., Efremova, M., Rieder, D.,
Hackl, H., and Trajanoski, Z. (2017). Pan-cancer Immunogenomic Analyses Finisguerra, V., Di Conza, G., Di Matteo, M., Serneels, J., Costa, S., Thompson,
Reveal Genotype-Immunophenotype Relationships and Predictors of Response A.A., Wauters, E., Walmsley, S., Prenen, H., Granot, Z., et al. (2015). MET is
to Checkpoint Blockade. Cell Rep. 18, 248–262. required for the recruitment of anti-tumoural neutrophils. Nature 522, 349–353.

Immunity 48, March 20, 2018 413


Immunity

Review
Fisher, G.H., Wellen, S.L., Klimstra, D., Lenczowski, J.M., Tichelaar, J.W., geting focal adhesion kinase renders pancreatic cancers responsive to check-
Lizak, M.J., Whitsett, J.A., Koretsky, A., and Varmus, H.E. (2001). Induction point immunotherapy. Nat. Med. 22, 851–860.
and apoptotic regression of lung adenocarcinomas by regulation of a K-Ras
transgene in the presence and absence of tumor suppressor genes. Genes Kang, T.W., Yevsa, T., Woller, N., Hoenicke, L., Wuestefeld, T., Dauch, D.,
Dev. 15, 3249–3262. Hohmeyer, A., Gereke, M., Rudalska, R., Potapova, A., et al. (2011). Senes-
cence surveillance of pre-malignant hepatocytes limits liver cancer develop-
Frederick, D.T., Piris, A., Cogdill, A.P., Cooper, Z.A., Lezcano, C., Ferrone, ment. Nature 479, 547–551.
C.R., Mitra, D., Boni, A., Newton, L.P., Liu, C., et al. (2013). BRAF inhibition
is associated with enhanced melanoma antigen expression and a more favor- Kastenhuber, E.R., and Lowe, S.W. (2017). Putting p53 in Context. Cell 170,
able tumor microenvironment in patients with metastatic melanoma. Clin. 1062–1078.
Cancer Res. 19, 1225–1231.
Katlinski, K.V., Gui, J., Katlinskaya, Y.V., Ortiz, A., Chakraborty, R., Bhatta-
Garon, E.B., Rizvi, N.A., Hui, R., Leighl, N., Balmanoukian, A.S., Eder, J.P., Pat- charya, S., Carbone, C.J., Beiting, D.P., Girondo, M.A., Peck, A.R., et al.
naik, A., Aggarwal, C., Gubens, M., Horn, L., et al.; KEYNOTE-001 Investiga- (2017). Inactivation of Interferon Receptor Promotes the Establishment of Im-
tors (2015). Pembrolizumab for the treatment of non-small-cell lung cancer. mune Privileged Tumor Microenvironment. Cancer Cell 31, 194–207.
N. Engl. J. Med. 372, 2018–2028.
Keck, M.K., Zuo, Z., Khattri, A., Stricker, T.P., Brown, C.D., Imanguli, M.,
Gentles, A.J., Newman, A.M., Liu, C.L., Bratman, S.V., Feng, W., Kim, D., Nair, €gelmann, J., et al. (2015). Integrative anal-
Rieke, D., Endhardt, K., Fang, P., Bra
V.S., Xu, Y., Khuong, A., Hoang, C.D., et al. (2015). The prognostic landscape ysis of head and neck cancer identifies two biologically distinct HPV and three
of genes and infiltrating immune cells across human cancers. Nat. Med. 21, non-HPV subtypes. Clin. Cancer Res. 21, 870–881.
938–945.
Kersten, K., de Visser, K.E., van Miltenburg, M.H., and Jonkers, J. (2017).
Glodde, N., Bald, T., van den Boorn-Konijnenberg, D., Nakamura, K., Genetically engineered mouse models in oncology research and cancer med-
O’Donnell, J.S., Szczepanski, S., Brandes, M., Eickhoff, S., Das, I., Shridhar, icine. EMBO Mol. Med. 9, 137–153.
N., et al. (2017). Reactive Neutrophil Responses Dependent on the Receptor
Knudson, A.G., Jr. (1971). Mutation and cancer: statistical study of retinoblas-
Tyrosine Kinase c-MET Limit Cancer Immunotherapy. Immunity 47, 789–
toma. Proc. Natl. Acad. Sci. USA 68, 820–823.
802 e789.
Kobielak, A., and Fuchs, E. (2006). Links between alpha-catenin, NF-kappaB,
Goel, S., DeCristo, M.J., Watt, A.C., BrinJones, H., Sceneay, J., Li, B.B., Khan,
and squamous cell carcinoma in skin. Proc. Natl. Acad. Sci. USA 103,
N., Ubellacker, J.M., Xie, S., Metzger-Filho, O., et al. (2017). CDK4/6 inhibition
2322–2327.
triggers anti-tumour immunity. Nature 548, 471–475.
Kortlever, R.M., Sodir, N.M., Wilson, C.H., Burkhart, D.L., Pellegrinet, L.,
Groenendijk, F.H., and Bernards, R. (2014). Drug resistance to targeted thera- Brown Swigart, L., Littlewood, T.D., and Evan, G.I. (2017). Myc Cooperates
pies: déjà vu all over again. Mol. Oncol. 8, 1067–1083. with Ras by Programming Inflammation and Immune Suppression. Cell 171,
1301–1315.e14.
Gunderson, A.J., Kaneda, M.M., Tsujikawa, T., Nguyen, A.V., Affara, N.I., Ruf-
fell, B., Gorjestani, S., Liudahl, S.M., Truitt, M., Olson, P., et al. (2016). Bruton Koya, R.C., Mok, S., Otte, N., Blacketor, K.J., Comin-Anduix, B., Tumeh, P.C.,
Tyrosine Kinase-Dependent Immune Cell Cross-talk Drives Pancreas Cancer. Minasyan, A., Graham, N.A., Graeber, T.G., Chodon, T., and Ribas, A. (2012).
Cancer Discov. 6, 270–285. BRAF inhibitor vemurafenib improves the antitumor activity of adoptive cell
immunotherapy. Cancer Res. 72, 3928–3937.
Hanahan, D., Wagner, E.F., and Palmiter, R.D. (2007). The origins of oncomice:
a history of the first transgenic mice genetically engineered to develop cancer. Koyama, S., Akbay, E.A., Li, Y.Y., Aref, A.R., Skoulidis, F., Herter-Sprie, G.S.,
Genes Dev. 21, 2258–2270. Buczkowski, K.A., Liu, Y., Awad, M.M., Denning, W.L., et al. (2016). STK11/
LKB1 Deficiency Promotes Neutrophil Recruitment and Proinflammatory
Herranz, N., Gallage, S., Mellone, M., Wuestefeld, T., Klotz, S., Hanley, C.J., Cytokine Production to Suppress T-cell Activity in the Lung Tumor Microenvi-
Raguz, S., Acosta, J.C., Innes, A.J., Banito, A., et al. (2015). mTOR regulates ronment. Cancer Res. 76, 999–1008.
MAPKAPK2 translation to control the senescence-associated secretory
phenotype. Nat. Cell Biol. 17, 1205–1217. Laberge, R.M., Sun, Y., Orjalo, A.V., Patil, C.K., Freund, A., Zhou, L., Curran,
S.C., Davalos, A.R., Wilson-Edell, K.A., Liu, S., et al. (2015). MTOR regulates
Hoare, M., Ito, Y., Kang, T.W., Weekes, M.P., Matheson, N.J., Patten, D.A., the pro-tumorigenic senescence-associated secretory phenotype by promot-
Shetty, S., Parry, A.J., Menon, S., Salama, R., et al. (2016). NOTCH1 mediates ing IL1A translation. Nat. Cell Biol. 17, 1049–1061.
a switch between two distinct secretomes during senescence. Nat. Cell Biol.
18, 979–992. Layer, J.P., Kronmu €ller, M.T., Quast, T., van den Boorn-Konijnenberg, D., Ef-
fern, M., Hinze, D., Althoff, K., Schramm, A., Westermann, F., Peifer, M.,
Hu-Lieskovan, S., Mok, S., Homet Moreno, B., Tsoi, J., Robert, L., Goedert, L., et al. (2017). Amplification of N-Myc is associated with a T-cell-poor microen-
Pinheiro, E.M., Koya, R.C., Graeber, T.G., Comin-Anduix, B., and Ribas, A. vironment in metastatic neuroblastoma restraining interferon pathway activity
(2015). Improved antitumor activity of immunotherapy with BRAF and MEK in- and chemokine expression. OncoImmunology 6, e1320626.
hibitors in BRAF(V600E) melanoma. Sci. Transl. Med. 7, 279ra41.
Le, D.T., Uram, J.N., Wang, H., Bartlett, B.R., Kemberling, H., Eyring, A.D.,
Hugo, W., Zaretsky, J.M., Sun, L., Song, C., Moreno, B.H., Hu-Lieskovan, S., Skora, A.D., Luber, B.S., Azad, N.S., Laheru, D., et al. (2015). PD-1 Blockade
Berent-Maoz, B., Pang, J., Chmielowski, B., Cherry, G., et al. (2016). Genomic in Tumors with Mismatch-Repair Deficiency. N. Engl. J. Med. 372, 2509–2520.
and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic
Melanoma. Cell 165, 35–44. Lennerz, V., Fatho, M., Gentilini, C., Frye, R.A., Lifke, A., Ferel, D., Wölfel, C.,
Huber, C., and Wölfel, T. (2005). The response of autologous T cells to a human
Huijbers, I.J. (2017). Generating Genetically Modified Mice: A Decision Guide. melanoma is dominated by mutated neoantigens. Proc. Natl. Acad. Sci. USA
Methods Mol. Biol. 1642, 1–19. 102, 16013–16018.

Jain, M., Arvanitis, C., Chu, K., Dewey, W., Leonhardt, E., Trinh, M., Sundberg, Lesina, M., Wörmann, S.M., Morton, J., Diakopoulos, K.N., Korneeva, O.,
C.D., Bishop, J.M., and Felsher, D.W. (2002). Sustained loss of a neoplastic Wimmer, M., Einwa €chter, H., Sperveslage, J., Demir, I.E., Kehl, T., et al.
phenotype by brief inactivation of MYC. Science 297, 102–104. (2016). RelA regulates CXCL1/CXCR2-dependent oncogene-induced senes-
cence in murine Kras-driven pancreatic carcinogenesis. J. Clin. Invest. 126,
Ji, H., Houghton, A.M., Mariani, T.J., Perera, S., Kim, C.B., Padera, R., Tonon, 2919–2932.
G., McNamara, K., Marconcini, L.A., Hezel, A., et al. (2006). K-ras activation
generates an inflammatory response in lung tumors. Oncogene 25, Linnemann, C., van Buuren, M.M., Bies, L., Verdegaal, E.M., Schotte, R., Calis,
2105–2112. J.J., Behjati, S., Velds, A., Hilkmann, H., Atmioui, D.E., et al. (2015). High-
throughput epitope discovery reveals frequent recognition of neo-antigens
Jiang, D., Gao, Z., Cai, Z., Wang, M., and He, J. (2015). Clinicopathological and by CD4+ T cells in human melanoma. Nat. Med. 21, 81–85.
prognostic significance of FOXP3+ tumor infiltrating lymphocytes in patients
with breast cancer: a meta-analysis. BMC Cancer 15, 727. Liu, F., Lang, R., Zhao, J., Zhang, X., Pringle, G.A., Fan, Y., Yin, D., Gu, F., Yao,
Z., and Fu, L. (2011). CD8+ cytotoxic T cell and FOXP3+ regulatory T cell infil-
Jiang, H., Hegde, S., Knolhoff, B.L., Zhu, Y., Herndon, J.M., Meyer, M.A., tration in relation to breast cancer survival and molecular subtypes. Breast
Nywening, T.M., Hawkins, W.G., Shapiro, I.M., Weaver, D.T., et al. (2016). Tar- Cancer Res. Treat. 130, 645–655.

414 Immunity 48, March 20, 2018


Immunity

Review
Lujambio, A., Akkari, L., Simon, J., Grace, D., Tschaharganeh, D.F., Bolden, Sarkar, S., Bristow, C.A., Dey, P., Rai, K., Perets, R., Ramirez-Cardenas, A.,
J.E., Zhao, Z., Thapar, V., Joyce, J.A., Krizhanovsky, V., and Lowe, S.W. Malasi, S., Huang-Hobbs, E., Haemmerle, M., Wu, S.Y., et al. (2017).
(2013). Non-cell-autonomous tumor suppression by p53. Cell 153, 449–460. PRKCI promotes immune suppression in ovarian cancer. Genes Dev. 31,
1109–1121.
Medrek, C., Pontén, F., Jirström, K., and Leandersson, K. (2012). The presence
of tumor associated macrophages in tumor stroma as a prognostic marker for Savas, P., Salgado, R., Denkert, C., Sotiriou, C., Darcy, P.K., Smyth, M.J., and
breast cancer patients. BMC Cancer 12, 306. Loi, S. (2016). Clinical relevance of host immunity in breast cancer: from TILs to
the clinic. Nat. Rev. Clin. Oncol. 13, 228–241.
Meylan, E., Dooley, A.L., Feldser, D.M., Shen, L., Turk, E., Ouyang, C., and
Jacks, T. (2009). Requirement for NF-kappaB signalling in a mouse model of Schumacher, T.N., and Schreiber, R.D. (2015). Neoantigens in cancer immu-
lung adenocarcinoma. Nature 462, 104–107. notherapy. Science 348, 69–74.

Moody, S.E., Sarkisian, C.J., Hahn, K.T., Gunther, E.J., Pickup, S., Dugan, Schuster, C., Berger, A., Hoelzl, M.A., Putz, E.M., Frenzel, A., Simma, O.,
K.D., Innocent, N., Cardiff, R.D., Schnall, M.D., and Chodosh, L.A. (2002). Con- Moritz, N., Hoelbl, A., Kovacic, B., Freissmuth, M., et al. (2011). The cooperat-
ditional activation of Neu in the mammary epithelium of transgenic mice results ing mutation or ‘‘second hit’’ determines the immunologic visibility toward
in reversible pulmonary metastasis. Cancer Cell 2, 451–461. MYC-induced murine lymphomas. Blood 118, 4635–4645.

Muller, P.A., and Vousden, K.H. (2014). Mutant p53 in cancer: new functions Schwitalla, S., Ziegler, P.K., Horst, D., Becker, V., Kerle, I., Begus-Nahrmann,
and therapeutic opportunities. Cancer Cell 25, 304–317. Y., Lechel, A., Rudolph, K.L., Langer, R., Slotta-Huspenina, J., et al. (2013).
Loss of p53 in enterocytes generates an inflammatory microenvironment
Muñoz-Espı́n, D., and Serrano, M. (2014). Cellular senescence: from physi- enabling invasion and lymph node metastasis of carcinogen-induced colo-
ology to pathology. Nat. Rev. Mol. Cell Biol. 15, 482–496. rectal tumors. Cancer Cell 23, 93–106.

Muñoz-Fontela, C., Mandinova, A., Aaronson, S.A., and Lee, S.W. (2016). Shchors, K., Shchors, E., Rostker, F., Lawlor, E.R., Brown-Swigart, L., and
Emerging roles of p53 and other tumour-suppressor genes in immune regula- Evan, G.I. (2006). The Myc-dependent angiogenic switch in tumors is medi-
tion. Nat. Rev. Immunol. 16, 741–750. ated by interleukin 1beta. Genes Dev. 20, 2527–2538.

Ock, C.Y., Hwang, J.E., Keam, B., Kim, S.B., Shim, J.J., Jang, H.J., Park, S., Shen, Q., Cohen, B., Zheng, W., Rahbar, R., Martin, B., Murakami, K., Lamorte,
Sohn, B.H., Cha, M., Ajani, J.A., et al. (2017). Genomic landscape associated S., Thompson, P., Berman, H., Zúñiga-Pflu €cker, J.C., et al. (2017). Notch
with potential response to anti-CTLA-4 treatment in cancers. Nat. Commun. Shapes the Innate Immunophenotype in Breast Cancer. Cancer Discov. 7,
8, 1050. 1320–1335.

Parker, J.S., Mullins, M., Cheang, M.C., Leung, S., Voduc, D., Vickery, T., Shin, D.S., Zaretsky, J.M., Escuin-Ordinas, H., Garcia-Diaz, A., Hu-Lieskovan,
Davies, S., Fauron, C., He, X., Hu, Z., et al. (2009). Supervised risk predictor S., Kalbasi, A., Grasso, C.S., Hugo, W., Sandoval, S., Torrejon, D.Y., et al.
of breast cancer based on intrinsic subtypes. J. Clin. Oncol. 27, 1160–1167. (2017). Primary Resistance to PD-1 Blockade Mediated by JAK1/2 Mutations.
Cancer Discov. 7, 188–201.
Peng, W., Chen, J.Q., Liu, C., Malu, S., Creasy, C., Tetzlaff, M.T., Xu, C.,
McKenzie, J.A., Zhang, C., Liang, X., et al. (2016). Loss of PTEN Promotes Sodir, N.M., Swigart, L.B., Karnezis, A.N., Hanahan, D., Evan, G.I., and Sou-
Resistance to T Cell-Mediated Immunotherapy. Cancer Discov. 6, 202–216. cek, L. (2011). Endogenous Myc maintains the tumor microenvironment.
Genes Dev. 25, 907–916.
Pérez-Mancera, P.A., Young, A.R., and Narita, M. (2014). Inside and out: the
activities of senescence in cancer. Nat. Rev. Cancer 14, 547–558. Soucek, L., Lawlor, E.R., Soto, D., Shchors, K., Swigart, L.B., and Evan, G.I.
(2007). Mast cells are required for angiogenesis and macroscopic expansion
Pribluda, A., Elyada, E., Wiener, Z., Hamza, H., Goldstein, R.E., Biton, M., Bur- of Myc-induced pancreatic islet tumors. Nat. Med. 13, 1211–1218.
stain, I., Morgenstern, Y., Brachya, G., Billauer, H., et al. (2013). A senescence-
inflammatory switch from cancer-inhibitory to cancer-promoting mechanism. Sparmann, A., and Bar-Sagi, D. (2004). Ras-induced interleukin-8 expression
Cancer Cell 24, 242–256. plays a critical role in tumor growth and angiogenesis. Cancer Cell 6, 447–458.

Pylayeva-Gupta, Y., Lee, K.E., Hajdu, C.H., Miller, G., and Bar-Sagi, D. (2012). Spranger, S., Bao, R., and Gajewski, T.F. (2015). Melanoma-intrinsic b-catenin
Oncogenic Kras-induced GM-CSF production promotes the development of signalling prevents anti-tumour immunity. Nature 523, 231–235.
pancreatic neoplasia. Cancer Cell 21, 836–847.
Spranger, S., Luke, J.J., Bao, R., Zha, Y., Hernandez, K.M., Li, Y., Gajewski,
Quigley, D., Silwal-Pandit, L., Dannenfelser, R., Langerød, A., Vollan, H.K., A.P., Andrade, J., and Gajewski, T.F. (2016). Density of immunogenic antigens
Vaske, C., Siegel, J.U., Troyanskaya, O., Chin, S.F., Caldas, C., et al. (2015). does not explain the presence or absence of the T-cell-inflamed tumor micro-
Lymphocyte Invasion in IC10/Basal-Like Breast Tumors Is Associated with environment in melanoma. Proc. Natl. Acad. Sci. USA 113, E7759–E7768.
Wild-Type TP53. Mol. Cancer Res. 13, 493–501.
Stanton, S.E., Adams, S., and Disis, M.L. (2016). Variation in the Incidence and
Rizvi, N.A., Hellmann, M.D., Snyder, A., Kvistborg, P., Makarov, V., Havel, J.J., Magnitude of Tumor-Infiltrating Lymphocytes in Breast Cancer Subtypes:
Lee, W., Yuan, J., Wong, P., Ho, T.S., et al. (2015). Cancer immunology. Muta- A Systematic Review. JAMA Oncol. 2, 1354–1360.
tional landscape determines sensitivity to PD-1 blockade in non-small cell lung
cancer. Science 348, 124–128. Stehelin, D., Varmus, H.E., Bishop, J.M., and Vogt, P.K. (1976). DNA related to
the transforming gene(s) of avian sarcoma viruses is present in normal avian
Robbins, P.F., Lu, Y.C., El-Gamil, M., Li, Y.F., Gross, C., Gartner, J., Lin, J.C., DNA. Nature 260, 170–173.
Teer, J.K., Cliften, P., Tycksen, E., et al. (2013). Mining exomic sequencing
data to identify mutated antigens recognized by adoptively transferred tu- Stewart, T.A., Pattengale, P.K., and Leder, P. (1984). Spontaneous mammary
mor-reactive T cells. Nat. Med. 19, 747–752. adenocarcinomas in transgenic mice that carry and express MTV/myc fusion
genes. Cell 38, 627–637.
Robinson, D.R., Wu, Y.M., Lonigro, R.J., Vats, P., Cobain, E., Everett, J., Cao,
X., Rabban, E., Kumar-Sinha, C., Raymond, V., et al. (2017). Integrative clinical Stiff, A., Trikha, P., Wesolowski, R., Kendra, K., Hsu, V., Uppati, S., McMichael,
genomics of metastatic cancer. Nature 548, 297–303. E., Duggan, M., Campbell, A., Keller, K., et al. (2016). Myeloid-Derived Sup-
pressor Cells Express Bruton’s Tyrosine Kinase and Can Be Depleted in Tu-
Rooney, M.S., Shukla, S.A., Wu, C.J., Getz, G., and Hacohen, N. (2015). mor-Bearing Hosts by Ibrutinib Treatment. Cancer Res. 76, 2125–2136.
Molecular and genetic properties of tumors associated with local immune
cytolytic activity. Cell 160, 48–61. Stodden, G.R., Lindberg, M.E., King, M.L., Paquet, M., MacLean, J.A., Mann,
J.L., DeMayo, F.J., Lydon, J.P., and Hayashi, K. (2015). Loss of Cdh1 and
Rous, P. (1911). A Sarcoma of the Fowl Transmissible by an Agent Separable Trp53 in the uterus induces chronic inflammation with modification of tumor
from the Tumor Cells. J. Exp. Med. 13, 397–411. microenvironment. Oncogene 34, 2471–2482.

Sagiv-Barfi, I., Kohrt, H.E., Czerwinski, D.K., Ng, P.P., Chang, B.Y., and Levy, Topper, M.J., Vaz, M., Chiappinelli, K.B., DeStefano Shields, C.E., Niknafs, N.,
R. (2015). Therapeutic antitumor immunity by checkpoint blockade is Yen, R.C., Wenzel, A., Hicks, J., Ballew, M., Stone, M., et al. (2017). Epigenetic
enhanced by ibrutinib, an inhibitor of both BTK and ITK. Proc. Natl. Acad. Therapy Ties MYC Depletion to Reversing Immune Evasion and Treating Lung
Sci. USA 112, E966–E972. Cancer. Cell 171, 1284–1300 e1221.

Immunity 48, March 20, 2018 415


Immunity

Review
Toso, A., Revandkar, A., Di Mitri, D., Guccini, I., Proietti, M., Sarti, M., Pinton, myeloid-derived suppressor cells to promote tumour initiation. Nat. Cell Biol.
S., Zhang, J., Kalathur, M., Civenni, G., et al. (2014). Enhancing chemotherapy 18, 632–644.
efficacy in Pten-deficient prostate tumors by activating the senescence-asso-
ciated antitumor immunity. Cell Rep. 9, 75–89. Wislez, M., Fujimoto, N., Izzo, J.G., Hanna, A.E., Cody, D.D., Langley, R.R.,
Tang, H., Burdick, M.D., Sato, M., Minna, J.D., et al. (2006). High expression
Van Allen, E.M., Miao, D., Schilling, B., Shukla, S.A., Blank, C., Zimmer, L., of ligands for chemokine receptor CXCR2 in alveolar epithelial neoplasia
Sucker, A., Hillen, U., Foppen, M.H.G., Goldinger, S.M., et al. (2015). Genomic induced by oncogenic kras. Cancer Res. 66, 4198–4207.
correlates of response to CTLA-4 blockade in metastatic melanoma. Science
350, 207–211. Wölfel, T., Hauer, M., Schneider, J., Serrano, M., Wölfel, C., Klehmann-Hieb,
E., De Plaen, E., Hankeln, T., Meyer zum Bu €schenfelde, K.H., and Beach, D.
van Rooij, N., van Buuren, M.M., Philips, D., Velds, A., Toebes, M., Heemskerk,
(1995). A p16INK4a-insensitive CDK4 mutant targeted by cytolytic T lympho-
B., van Dijk, L.J., Behjati, S., Hilkmann, H., El Atmioui, D., et al. (2013). Tumor
cytes in a human melanoma. Science 269, 1281–1284.
exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-
responsive melanoma. J. Clin. Oncol. 31, e439–e442.
Wörmann, S.M., Song, L., Ai, J., Diakopoulos, K.N., Kurkowski, M.U.,
Vanden Borre, P., Gunda, V., McFadden, D.G., Sadow, P.M., Varmeh, S., Ber- € lu
Görgu €, K., Ruess, D., Campbell, A., Doglioni, C., Jodrell, D., et al.
nasconi, M., and Parangi, S. (2014). Combined BRAF(V600E)- and SRC-inhibi- (2016). Loss of P53 Function Activates JAK2-STAT3 Signaling to Promote
tion induces apoptosis, evokes an immune response and reduces tumor Pancreatic Tumor Growth, Stroma Modification, and Gemcitabine Resis-
growth in an immunocompetent orthotopic mouse model of anaplastic thyroid tance in Mice and Is Associated With Patient Survival. Gastroenterology
cancer. Oncotarget 5, 3996–4010. 151, 180–193.e12.

Ventura, A., Kirsch, D.G., McLaughlin, M.E., Tuveson, D.A., Grimm, J., Lintault, Xue, W., Zender, L., Miething, C., Dickins, R.A., Hernando, E., Krizhanovsky,
L., Newman, J., Reczek, E.E., Weissleder, R., and Jacks, T. (2007). Restoration V., Cordon-Cardo, C., and Lowe, S.W. (2007). Senescence and tumour clear-
of p53 function leads to tumour regression in vivo. Nature 445, 661–665. ance is triggered by p53 restoration in murine liver carcinomas. Nature 445,
656–660.
von Hansemann, D. (1890). Ueber asymmetrische Zelltheilung in Epithelkreb-
sen und deren biologische Bedeutung. Virchows Arch. Path. Anat 119, Yang, Y. (2015). Cancer immunotherapy: harnessing the immune system to
299–326. battle cancer. J. Clin. Invest. 125, 3335–3337.
Wang, G., Lu, X., Dey, P., Deng, P., Wu, C.C., Jiang, S., Fang, Z., Zhao, K., Ko-
naparthi, R., Hua, S., et al. (2016). Targeting YAP-Dependent MDSC Infiltration Yetil, A., Anchang, B., Gouw, A.M., Adam, S.J., Zabuawala, T., Parameswaran,
Impairs Tumor Progression. Cancer Discov. 6, 80–95. R., van Riggelen, J., Plevritis, S., and Felsher, D.W. (2015). p19ARF is a critical
mediator of both cellular senescence and an innate immune response associ-
Wang, Q., Hu, B., Hu, X., Kim, H., Squatrito, M., Scarpace, L., deCarvalho, ated with MYC inactivation in mouse model of acute leukemia. Oncotarget 6,
A.C., Lyu, S., Li, P., Li, Y., et al. (2017). Tumor Evolution of Glioma-Intrinsic 3563–3577.
Gene Expression Subtypes Associates with Immunological Changes in the
Microenvironment. Cancer Cell 32, 42–56 e46. Ying, H., Elpek, K.G., Vinjamoori, A., Zimmerman, S.M., Chu, G.C., Yan, H.,
Fletcher-Sananikone, E., Zhang, H., Liu, Y., Wang, W., et al. (2011). PTEN is
Welte, T., Kim, I.S., Tian, L., Gao, X., Wang, H., Li, J., Holdman, X.B., Hersch- a major tumor suppressor in pancreatic ductal adenocarcinoma and regulates
kowitz, J.I., Pond, A., Xie, G., et al. (2016). Oncogenic mTOR signalling recruits an NF-kB-cytokine network. Cancer Discov. 1, 158–169.

416 Immunity 48, March 20, 2018

You might also like